1
|
Travaglio Morales D, Coronado Poggio M, Huerga Cabrerizo C, Losantos García I, Escabias del Pozo C, Lancha Hernández C, Rodado Marina S, Domínguez Gadea L. Prognostic Value of Pretreatment 18F-FDG-PET/CT Metabolic Parameters in Advanced High-Grade Serous Ovarian Cancer. Cancers (Basel) 2025; 17:698. [PMID: 40002291 PMCID: PMC11853401 DOI: 10.3390/cancers17040698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
AIM To assess the prognostic value of pretreatment 18F-FDG-PET/CT quantitative metabolic parameters in patients with advanced high-grade serous ovarian cancer (HGSOC). METHODS A review of 47 patients diagnosed with advanced HGSOC between 2012 and 2020 in our center was performed, evaluating pretreatment 18F-FDG-PET/CT metabolic parameters: maximum standardized uptake value (SUVmax), total lesion glycolysis (TLG) and metabolic tumoral volume (MTV). Two experienced nuclear medicine physicians evaluated the images, thereby obtaining quantitative parameters semiautomatically classifying the volume of interest (VOI) as the target (t): VOI with the highest SUVmax normalized by lean body mass (SUVmax(lbm)), non target (nt) and total (sum of target and non-target VOIs). The disease-free survival (DFS) and overall survival (OS) were calculated. Optimal cutoff values with ROC curves/median values were used. The Correlation between metabolic parameters and DFS/OS was determined using univariate and survival-curves analysis. RESULTS The median DFS was 18 months (2.5-55) and the OS 33.6 months (2.5-92). The MTVtotal, MTV(t), TLGtotal and TLG(t) were significantly associated with DFS (p = 0.005, 0.01, 0.04 and 0.04, respectively). The patients with MTVtotal > 427.8 cm3 and MTVtarget > 434 cm3 had shorter DFS than the patients with lower values (18.8 versus 31 months and 15.6 versus 30, p = 0.02 and 0.01, respectively). The patients with higher TLGtotal and TLG(t) values tended to have worse DFS (p = 0.26 and 0.31, respectively). In a multivariate analysis, the MTVtotal was statistically significantly associated with DFS (p = 0.003). No correlation was found with OS. CONCLUSIONS Pretreatment MTVtotal and MTV(t) appear to be predictive of relapse in patients with advanced HGSOC.
Collapse
Affiliation(s)
- Daniela Travaglio Morales
- Nuclear Medicine Department, La Paz University Hospital, 28046 Madrid, Spain
- Doctoral School, Universidad Autónoma of Madrid, 28049 Madrid, Spain
- Nuclear Medicine Department, Leipzig University Hospital, 04103 Leipzig, Germany
| | | | | | | | | | | | - Sonia Rodado Marina
- Nuclear Medicine Department, La Paz University Hospital, 28046 Madrid, Spain
| | | |
Collapse
|
2
|
Egi R, Matsusaka Y, Watanabe K, Seto A, Matsunari I, Arai T, Nakano S, Kuji I. Single-center analysis of cardiac amyloidosis using 99m Tc-HMDP imaging for diagnosis and evaluation after tafamidis treatment. Nucl Med Commun 2025; 46:38-46. [PMID: 39483085 PMCID: PMC11634134 DOI: 10.1097/mnm.0000000000001922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE This study aimed to evaluate the diagnostic performance of 99m Tc-hydroxymethylene diphosphonate ( 99m Tc-HMDP) imaging for cardiac amyloidosis and to demonstrate changes in cardiac uptake of 99m Tc-HMDP after tafamidis treatment. METHODS Seventy-five patients with suspected cardiac amyloidosis who underwent 99m Tc-HMDP imaging were included. We compared visual Perugini grades and semiquantitative heart-to-contralateral (H/CL) area ratios, myocardial maximum standardized uptake value (SUVmax), and peak of SUV (SUVpeak) between cardiac transthyretin amyloidosis (ATTR) and amyloid light-chain amyloidosis (AL). Comparison of interobserver reproducibility between H/CL ratios and myocardial SUVmax/SUVpeak was performed. H/CL ratio of 99m Tc-HMDP and myocardial SUVmax/SUVpeak were compared before and after tafamidis administration for cardiac wild-type ATTR. RESULTS Among 75 patients, 20 patients (26.7%) were visually positive based on Perugini grade. Fifteen and three patients were pathologically identified as cardiac ATTR and AL, respectively. ATTR group ( n = 15) had significantly higher H/CL ratios of 99m Tc-HMDP than AL group ( n = 3) ( P = 0.003). ATTR group ( n = 15) had significantly higher myocardial SUVmax/SUVpeak of 99m Tc-HMDP than AL group ( n = 2) ( P = 0.015). Myocardial SUVmax/SUVpeak had better interobserver reproducibility than H/CL ratios. After tafamidis treatment for cardiac wild-type ATTR, the decrease in myocardial SUVpeak was significant but not in H/CL ratios and myocardial SUVmax. CONCLUSION H/CL ratio and SUVmax/SUVpeak in 99m Tc-HMDP imaging were useful for diagnosing cardiac ATTR. Myocardial SUVpeak may be useful for monitoring changes in cardiac uptake after tafamidis treatment for cardiac ATTR.
Collapse
Affiliation(s)
- Ryuta Egi
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Saitama
| | - Yohji Matsusaka
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Saitama
| | - Kaho Watanabe
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Saitama
| | - Akira Seto
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Saitama
| | - Ichiro Matsunari
- Division of Nuclear Medicine, Department of Radiology, Saitama Medical University Hospital, Moroyama
| | - Takahide Arai
- Department of Cardiology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Shintaro Nakano
- Department of Cardiology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Saitama
| |
Collapse
|
3
|
Guo R, Yan W, Wang F, Su H, Meng X, Xie Q, Zhao W, Yang Z, Li N. The utility of 18F-FDG PET/CT for predicting the pathological response and prognosis to neoadjuvant immunochemotherapy in resectable non-small-cell lung cancer. Cancer Imaging 2024; 24:120. [PMID: 39256860 PMCID: PMC11385245 DOI: 10.1186/s40644-024-00772-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
OBJECTIVE To evaluate the potential utility of 18F-FDG PET/CT to assess response to neoadjuvant immunochemotherapy in patients with resectable NSCLC, and the ability to screen patients who may benefit from neoadjuvant immunochemotherapy. METHODS Fifty one resectable NSCLC (stage IA-IIIB) patients were analyzed, who received two-three cycles neoadjuvant immunochemotherapy.18F-FDG PET/CT was carried out at baseline(scan-1) and prior to radical resection(scan-2). SULmax, SULpeak, MTV, TLG, T/N ratio, ΔSULmax%,ΔSULpeak%, ΔMTV%, ΔTLG%,ΔT/N ratio% were calculated. 18F-FDG PET/CT responses were classified using PERCIST. We then compared the RECIST 1.1 and PERCIST criteria for response assessment.With surgical pathology of primary lesions as the gold standard, the correlation between metabolic parameters of 18F-FDG PET/CT and major pathologic response (MPR) was analyzed. All metabolic parameters were compared to treatment response and correlated to PFS and OS. RESULTS In total of fifty one patients, MPR was achieved in 25(49%, 25/51) patients after neoadjuvant therapy. The metabolic parameters of Scan-1 were not correlated with MPR.The degree of pathological regression was negatively correlated with SULmax, SULpeak, MTV, TLG, T/N ratio of scan-2, and the percentage changes of the ΔSULmax%, ΔSULpeak%, ΔMTV%,ΔTLG%,ΔT/N ratio% after neoadjuvant therapy (p < 0.05). According to PERCIST, 36 patients (70.6%, 36/51) showed PMR, 12 patients(23.5%, 12/51) had stable metabolic disease(SMD), and 3 patients(5.9%, 3/51) had progressive metabolic disease (PMD). ROC indicated that all of scan-2 metabolic parameters and the percentage changes of metabolic parameters had ability to predict MPR and non-MPR, SULmax and T/N ratio of scan-2 had the best differentiation ability.The accuracy of RECIST 1.1 and PERCIST criteria were no statistical significance(p = 0.91). On univariate analysis, ΔMTV% has the highest correlation with PFS. CONCLUSIONS Metabolic response by 18F-FDG PET/CT can predict MPR to neoadjuvant immunochemotherapy in resectable NSCLC. ΔMTV% was significantly correlated with PFS.
Collapse
Affiliation(s)
- Rui Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wanpu Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery I, Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Fei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Qing Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
4
|
Ince S, Laforest R, Itani M, Prasad V, Derenoncourt PR, Crandall JP, Ashrafinia S, Smith AM, Wahl RL, Fraum TJ. Quantitative Assessments of Tumor Activity in a General Oncologic PET/CT Population: Which Metric Minimizes Tracer Uptake Time Dependence? J Nucl Med 2024; 65:1349-1356. [PMID: 39142828 PMCID: PMC11372261 DOI: 10.2967/jnumed.123.266469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/10/2024] [Indexed: 08/16/2024] Open
Abstract
In oncologic PET, the SUV and standardized uptake ratio (SUR) of a viable tumor generally increase during the postinjection period. In contrast, the net influx rate (Ki ), which is derived from dynamic PET data, should remain relatively constant. Uptake-time-corrected SUV (cSUV) and SUR (cSUR) have been proposed as uptake-time-independent, static alternatives to Ki Our primary aim was to quantify the intrascan repeatability of Ki , SUV, cSUV, SUR, and cSUR among malignant lesions on PET/CT. An exploratory aim was to assess the ability of cSUR to estimate Ki Methods: This prospective, single-center study enrolled adults undergoing standard-of-care oncologic PET/CT. SUV and Ki images were reconstructed from dynamic PET data obtained before (∼35-50 min after injection) and after (∼75-90 min after injection) standard-of-care imaging. Tumors were manually segmented. Quantitative metrics were extracted. cSUVs and cSURs were calculated for a 60-min postinjection reference uptake time. The magnitude of the intrascan test-retest percent change (test-retest |%Δ|) was calculated. Coefficients of determination (R 2) and intraclass correlation coefficients (ICC) were also computed. Differences between metrics were assessed via the Wilcoxon signed-rank test (α, 0.05). Results: This study enrolled 78 subjects; 41 subjects (mean age, 63.8 y; 24 men) with 116 lesions were analyzed. For both tracers, SUVmax and maximum SUR (SURmax) had large early-to-late increases (i.e., poor intrascan repeatability). Among [18F]FDG-avid lesions (n = 93), there were no differences in intrascan repeatability (median test-retest |%Δ|; ICC) between the maximum Ki (Ki ,max) (13%; 0.97) and either the maximum cSUV (cSUVmax) (12%, P = 0.90; 0.96) or the maximum cSUR (cSURmax) (13%, P = 0.67; 0.94). For DOTATATE-avid lesions (n = 23), there were no differences in intrascan repeatability between the Ki ,max (11%; 0.98) and either the cSUVmax (13%, P = 0.41; 0.98) or the cSURmax (11%, P = 0.08; 0.94). The SUVmax, cSUVmax, SURmax, and cSURmax were all strongly correlated with the Ki ,max for both [18F]FDG (R 2, 0.81-0.92) and DOTATATE (R 2, 0.88-0.96), but the cSURmax provided the best agreement with the Ki ,max across early-to-late time points for [18F]FDG (ICC, 0.69-0.75) and DOTATATE (ICC, 0.90-0.91). Conclusion: Ki ,max, cSUVmax, and cSURmax had low uptake time dependence compared with SUVmax and SURmax The Ki ,max can be predicted from cSURmax.
Collapse
Affiliation(s)
- Semra Ince
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Richard Laforest
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Malak Itani
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Vikas Prasad
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - John P Crandall
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Anne M Smith
- Siemens Medical Solutions Inc., Knoxville, Tennessee; and
| | - Richard L Wahl
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Tyler J Fraum
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri;
| |
Collapse
|
5
|
Hashimoto F, Onishi Y, Ote K, Tashima H, Yamaya T. Two-step optimization for accelerating deep image prior-based PET image reconstruction. Radiol Phys Technol 2024; 17:776-781. [PMID: 39096446 DOI: 10.1007/s12194-024-00831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Deep learning, particularly convolutional neural networks (CNNs), has advanced positron emission tomography (PET) image reconstruction. However, it requires extensive, high-quality training datasets. Unsupervised learning methods, such as deep image prior (DIP), have shown promise for PET image reconstruction. Although DIP-based PET image reconstruction methods demonstrate superior performance, they involve highly time-consuming calculations. This study proposed a two-step optimization method to accelerate end-to-end DIP-based PET image reconstruction and improve PET image quality. The proposed two-step method comprised a pre-training step using conditional DIP denoising, followed by an end-to-end reconstruction step with fine-tuning. Evaluations using Monte Carlo simulation data demonstrated that the proposed two-step method significantly reduced the computation time and improved the image quality, thereby rendering it a practical and efficient approach for end-to-end DIP-based PET image reconstruction.
Collapse
Affiliation(s)
- Fumio Hashimoto
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamana-Ku, Hamamatsu, 434-8601, Japan.
- Graduate School of Science and Engineering, Chiba University, 1-33, Yayoicho,Inage-Ku, Chiba, 263-8522, Japan.
- National Institutes for Quantum Science and Technology, 4-9-1, Anagawa,Inage-Ku, Chiba, 263-8555, Japan.
| | - Yuya Onishi
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamana-Ku, Hamamatsu, 434-8601, Japan
| | - Kibo Ote
- Central Research Laboratory, Hamamatsu Photonics K. K, 5000 Hirakuchi, Hamana-Ku, Hamamatsu, 434-8601, Japan
| | - Hideaki Tashima
- National Institutes for Quantum Science and Technology, 4-9-1, Anagawa,Inage-Ku, Chiba, 263-8555, Japan
| | - Taiga Yamaya
- Graduate School of Science and Engineering, Chiba University, 1-33, Yayoicho,Inage-Ku, Chiba, 263-8522, Japan
- National Institutes for Quantum Science and Technology, 4-9-1, Anagawa,Inage-Ku, Chiba, 263-8555, Japan
| |
Collapse
|
6
|
Sun Y, Deng M, Gevaert O, Aberle M, Olde Damink SW, van Dijk D, Rensen SS. Tumor metabolic activity is associated with subcutaneous adipose tissue radiodensity and survival in non-small cell lung cancer. Clin Nutr 2024; 43:1809-1815. [PMID: 38870661 DOI: 10.1016/j.clnu.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Cachexia-associated body composition alterations and tumor metabolic activity are both associated with survival of cancer patients. Recently, subcutaneous adipose tissue properties have emerged as particularly prognostic body composition features. We hypothesized that tumors with higher metabolic activity instigate cachexia related peripheral metabolic alterations, and investigated whether tumor metabolic activity is associated with body composition and survival in patients with non-small-cell lung cancer (NSCLC), focusing on subcutaneous adipose tissue. METHODS A retrospective analysis was performed on a cohort of 173 patients with NSCLC. 18F-fluorodeoxyglucose positron emission tomography-computed tomography (PET-CT) scans obtained before treatment were used to analyze tumor metabolic activity (standardized uptake value (SUV) and SUV normalized by lean body mass (SUL)) as well as body composition variables (subcutaneous and visceral adipose tissue radiodensity (SAT/VAT radiodensity) and area; skeletal muscle radiodensity (SM radiodensity) and area). Subjects were divided into groups with high or low SAT radiodensity based on Youden Index of Receiver Operator Characteristics (ROC). Associations between tumor metabolic activity, body composition variables, and survival were analyzed by Mann-Whitney tests, Cox regression, and Kaplan-Meier analysis. RESULTS The overall prevalence of high SAT radiodensity was 50.9% (88/173). Patients with high SAT radiodensity had shorter survival compared with patients with low SAT radiodensity (mean: 45.3 vs. 50.5 months, p = 0.026). High SAT radiodensity was independently associated with shorter overall survival (multivariate Cox regression HR = 1.061, 95% CI: 1.022-1.101, p = 0.002). SAT radiodensity also correlated with tumor metabolic activity (SULpeak rs = 0.421, p = 0.029; SUVpeak rs = 0.370, p = 0.048). In contrast, the cross-sectional areas of SM, SAT, and VAT were not associated with tumor metabolic activity or survival. CONCLUSION Higher SAT radiodensity is associated with higher tumor metabolic activity and shorter survival in patients with NSCLC. This may suggest that tumors with higher metabolic activity induce subcutaneous adipose tissue alterations such as decreased lipid density, increased fibrosis, or browning.
Collapse
Affiliation(s)
- Yan Sun
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Min Deng
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, USA; Stanford Center for Biomedical Informatics Research, Department of Biomedical Data Science, Stanford University, USA
| | - Merel Aberle
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Steven W Olde Damink
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; Department of General, Visceral- and Transplantation Surgery, University Hospital Essen, Duisberg-Essen University, Germany
| | - David van Dijk
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Sander S Rensen
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
7
|
Yang T, Liu D, Zhang Z, Sa R, Guan F. Predicting T-Cell Lymphoma in Children From 18F-FDG PET-CT Imaging With Multiple Machine Learning Models. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:952-964. [PMID: 38321311 PMCID: PMC11169166 DOI: 10.1007/s10278-024-01007-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/08/2024]
Abstract
This study aimed to examine the feasibility of utilizing radiomics models derived from 18F-FDG PET/CT imaging to screen for T-cell lymphoma in children with lymphoma. All patients had undergone 18F-FDG PET/CT scans. Lesions were extracted from PET/CT and randomly divided into training and validation sets. Two different types of models were constructed as follows: features that are extracted from standardized uptake values (SUV)-associated parameters, and CT images were used to build SUV/CT-based model. Features that are derived from PET and CT images were used to build PET/CT-based model. Logistic regression (LR), linear support vector machine, support vector machine with the radial basis function kernel, neural networks, and adaptive boosting were performed as classifiers in each model. In the training sets, 77 patients, and 247 lesions were selected for building the models. In the validation sets, PET/CT-based model demonstrated better performance than that of SUV/CT-based model in the prediction of T-cell lymphoma. LR showed highest accuracy with 0.779 [0.697, 0.860], area under the receiver operating characteristic curve (AUC) with 0.863 [0.762, 0.963], and preferable goodness-of-fit in PET/CT-based model at the patient level. LR also showed best performance with accuracy of 0.838 [0.741, 0.936], AUC of 0.907 [0.839, 0.976], and preferable goodness-of-fit in PET/CT-based model at the lesion level. 18F-FDG PET/CT-based radiomics models with different machine learning classifiers were able to screen T-cell lymphoma in children with high accuracy, AUC, and preferable goodness-of-fit, providing incremental value compared with SUV-associated features.
Collapse
Affiliation(s)
- Taiyu Yang
- Department of Nuclear Medicine, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, China
| | - Danyan Liu
- Department of Radiology, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, China
| | - Zexu Zhang
- Department of Nuclear Medicine, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, China
| | - Ri Sa
- Department of Nuclear Medicine, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, China.
| | - Feng Guan
- Department of Nuclear Medicine, The First Hospital of Jilin University, 1# Xinmin St, Changchun, 130021, China.
| |
Collapse
|
8
|
Xu K, Kang H. A Review of Machine Learning Approaches for Brain Positron Emission Tomography Data Analysis. Nucl Med Mol Imaging 2024; 58:203-212. [PMID: 38932757 PMCID: PMC11196571 DOI: 10.1007/s13139-024-00845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 06/28/2024] Open
Abstract
Positron emission tomography (PET) imaging has moved forward the development of medical diagnostics and research across various domains, including cardiology, neurology, infection detection, and oncology. The integration of machine learning (ML) algorithms into PET data analysis has further enhanced their capabilities of including disease diagnosis and classification, image segmentation, and quantitative analysis. ML algorithms empower researchers and clinicians to extract valuable insights from complex big PET datasets, which enabling automated pattern recognition, predictive health outcome modeling, and more efficient data analysis. This review explains the basic knowledge of PET imaging, statistical methods for PET image analysis, and challenges of PET data analysis. We also discussed the improvement of analysis capabilities by combining PET data with machine learning algorithms and the application of this combination in various aspects of PET image research. This review also highlights current trends and future directions in PET imaging, emphasizing the driving and critical role of machine learning and big PET image data analytics in improving diagnostic accuracy and personalized medical approaches. Integration between PET imaging will shape the future of medical diagnosis and research.
Collapse
Affiliation(s)
- Ke Xu
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 1100, Nashville, TN 37203 USA
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 1100, Nashville, TN 37203 USA
| |
Collapse
|
9
|
Fu X, Cai Z, Fu S, Cai H, Li M, Gu H, Jin R, Xia C, Lui S, Song B, Gong Q, Ai H. Porphyrin-Based Self-Assembled Nanoparticles for PET/MR Imaging of Sentinel Lymph Node Metastasis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27139-27150. [PMID: 38752591 DOI: 10.1021/acsami.4c03611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Diagnosing of lymph node metastasis is challenging sometimes, and multimodal imaging offers a promising method to improve the accuracy. This work developed porphyrin-based nanoparticles (68Ga-F127-TAPP/TCPP(Mn) NPs) as PET/MR dual-modal probes for lymph node metastasis imaging by a simple self-assembly method. Compared with F127-TCPP(Mn) NPs, F127-TAPP/TCPP(Mn) NPs synthesized by amino-porphyrins (TAPP) doping can not only construct PET/MR bimodal probes but also improve the T1 relaxivity (up to 456%). Moreover, T1 relaxivity can be adjusted by altering the molar ratio of TAPP/TCPP(Mn) and the concentration of F127. However, a similar increase in T1 relaxivity was not observed in the F127-TCPP/TCPP(Mn) NPs, which were synthesized using carboxy-porphyrins (TCPP) doping. In a breast cancer lymph node metastasis mice model, subcutaneous injection of 68Ga-F127-TAPP/TCPP(Mn) NPs through the hind foot pad, the normal lymph nodes and metastatic lymph nodes were successfully distinguished based on the difference of PET standard uptake values and MR signal intensities. Furthermore, the dark brown F127-TAPP/TCPP(Mn) NPs demonstrated the potential for staining and mapping lymph nodes. This study provides valuable insights into developing and applying PET/MR probes for lymph node metastasis imaging.
Collapse
Affiliation(s)
- Xiaomin Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Shengxiang Fu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Huawei Cai
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mufeng Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haojie Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Su Lui
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Psychoradiology Research Unit of Chinese Academy of Medical Sciences, Sichuan University, Chengdu 610041, China
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Samimi R, Kamali-Asl A, Ahmadyar Y, van den Hoff J, Geramifar P, Rahmim A. Dual time-point [ 18F]FDG PET imaging for quantification of metabolic uptake rate: Evaluation of a simple, clinically feasible method. Phys Med 2024; 121:103336. [PMID: 38626637 DOI: 10.1016/j.ejmp.2024.103336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/18/2024] Open
Abstract
PURPOSE We aimed to investigate whether a clinically feasible dual time-point (DTP) approach can accurately estimate the metabolic uptake rate constant (Ki) and to explore reliable acquisition times through simulations and clinical assessment considering patient comfort and quantification accuracy. METHODS We simulated uptake kinetics in different tumors for four sets of DTP PET images within the routine clinical static acquisition at 60-min post-injection (p.i.). We determined Ki for a total of 81 lesions. Ki quantification from full dynamic PET data (Patlak-Ki) and Ki from DTP (DTP-Ki) were compared. In addition, we scaled a population-based input function (PBIFscl) with the image-derived blood pool activity sampled at different time points to assess the best scaling time-point for Ki quantifications in the simulation data. RESULTS In the simulation study, Ki estimated using DTP via (30,60-min), (30,90-min), (60,90-min), and (60,120-min) samples showed strong correlations (r ≥ 0.944, P < 0.0001) with the true value of Ki. The DTP results with the PBIFscl at 60-min time-point in (30,60-min), (60,90-min), and (60,120-min) were linearly related to the true Ki with a slope of 1.037, 1.008, 1.013 and intercept of -6 × 10-4, 2 × 10-5, 5 × 10-5, respectively. In a clinical study, strong correlations (r ≥ 0.833, P < 0.0001) were observed between Patlak-Ki and DTP-Ki. The Patlak-derived mean values of Ki, tumor-to-background-ratio, signal-to-noise-ratio, and contrast-to-noise-ratio were linearly correlated with the DTP method. CONCLUSIONS Besides calculating the retention index as a commonly used quantification parameter inDTP imaging,our DTP method can accurately estimate Ki.
Collapse
Affiliation(s)
- Rezvan Samimi
- Department of Radiation Medicine Engineering, Shahid Beheshti University, Tehran, Iran
| | - Alireza Kamali-Asl
- Department of Radiation Medicine Engineering, Shahid Beheshti University, Tehran, Iran.
| | - Yashar Ahmadyar
- Department of Radiation Medicine Engineering, Shahid Beheshti University, Tehran, Iran
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden 01328, Germany; Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arman Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| |
Collapse
|
11
|
Shirakawa Y, Matsutomo N, Suyama J. Feasibility of noise-reduction reconstruction technology based on non-local-mean principle in SiPM-PET/CT. Phys Med 2024; 119:103303. [PMID: 38325223 DOI: 10.1016/j.ejmp.2024.103303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/09/2023] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Quantitative values of positron emission tomography (PET) images using non-local-mean in a silicon photomultiplier (SiPM)-PET/computed tomography (CT) system with phantom and clinical images. The evaluation was conducted on a National Electrical Manufacturers Association body phantom with micro-spheres (4, 5, 6, 8, 10, 13 mm) and clinical images using the SiPM-PET/CT system. The signal-to-background ratio of the phantom was set to 4, and all PET image data was obtained and reconstructed using three-dimensional ordered subset expectation maximization, time-of-flight, point-spread function, and a 4-mm Gaussian filter (GF) and clear adaptive low-noise method (CaLM) in mild, standard, and strong intensities. The evaluation included the standardized uptake value (SUV), percent contrast (QH), coefficient of variation of the background area (CVbackground) clinical imaging for SUV of lung nodules, liver signal-to-noise ratio (SNR), and visual evaluation. SUVmax for 8-mm sphere in phantom images at 2 min for GF and CaLM (mild, standard, strong) were 2.11, 2.32, 2.02, and 1.72; the QH, 8 mm was 27.33 %, 27.47 %, 21.81 %, and 16.09 %; and CVbackground was 12.78, 11.35, 7.86, and 4.71, respectively. CaLM demonstrated higher SUVmax in clinical images than GF for all lung nodule sizes. The average SUVmax for nodules with a diameter of ≤ 1 cm were 5.9 ± 2.4, 9.9 ± 4.9, 9.9 ± 5.0, and 9.9 ± 5.0 for GF and CaLM-mild, standard, and strong intensities, respectively. Liver SNRs were higher for CaLM (mild, standard, strong) compared to GF, with increasing CaLM intensity causing higher liver SNR. CaLM-mild and standard demonstrated suitability for diagnosis in visual evaluation.
Collapse
Affiliation(s)
- Yuya Shirakawa
- Department of Radiology, Kyorin University Hospital, Tokyo, Japan.
| | - Norikazu Matsutomo
- Department of Medical Radiological Technology, Faculty of Health Sciences, Kyorin University, Japan.
| | - Jumpei Suyama
- Department of Radiology, Faculty of Medicine, Kyorin University, Tokyo, Japan.
| |
Collapse
|
12
|
Kennedy JA, Palchan-Hazan T, Maronnier Q, Caselles O, Courbon F, Levy M, Keidar Z. An extended bore length solid-state digital-BGO PET/CT system: design, preliminary experience, and performance characteristics. Eur J Nucl Med Mol Imaging 2024; 51:954-964. [PMID: 38012446 DOI: 10.1007/s00259-023-06514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE A solid-state PET/CT system uses bismuth germanium oxide (BGO) scintillating crystals coupled to silicon photomultipliers over an extended 32 cm axial field-of-view (FOV) to provide high spatial resolution and very high sensitivity. Performance characteristics were determined for this digital-BGO system, including NEMA and EARL standards. METHODS Spatial resolution, scatter fraction (SF), noise equivalent count rate (NECR), sensitivity, count rate accuracy, and image quality (IQ) were evaluated for the digital-BGO system as per NEMA NU 2-2018, at 2 sites of first clinical install. System energy resolution was measured. Bayesian penalized-likelihood reconstruction (BPL) was used for IQ. EARL Standards 2 studies were reconstructed by BPL combined with a contrast-enhancing deep learning algorithm. An Esser PET phantom was evaluated. Three patient examples were obtained with low-dose radiotracer activity: 2 MBq/kg of [18F]FDG ([18F]-2-fluoro-2-deoxy-D-glucose), 2.3 MBq/kg [68Ga]Ga-DOTA-TATE ([dodecane tetra-acetic acid,Tyr3]-octreotate), and 14.5 MBq/kg [82Rb]RbCl ([82Rb]-rubidium-chloride). Total scan times were ≤ 8 min. RESULTS NEMA sensitivity was 47.6 cps/kBq at the axial center. Spatial resolution at 1 cm from the center axis was ≤4.5 mm (filtered back projection) and ≤3.8 mm (ordered subset expectation maximization). SF was 35.6%, count rate accuracy was 2.16%, and peak NECR was 485.2 kcps at 16.9 kBq/mL. Contrast for IQ was 61.1 to 90.7% (smallest to largest sphere) with background variations from 7.6 to 2.1%, and a "lung" error of 4.7%. The average detector energy resolution was 9.67%. Image quality for patient scans was good. EARL Standards 2 criteria were robustly met and Esser phantom features ≥4.8 mm were resolved at 2 min per bed position. CONCLUSION A solid-state 32 cm axial FOV digital-BGO PET/CT system provides good spatial and energy resolution, high count rates, and superior NEMA sensitivity in its class, enabling fast clinical acquisitions with low-dose radiotracer activity.
Collapse
Affiliation(s)
- John A Kennedy
- Department of Nuclear Medicine, Rambam Health Care Campus, P.O.B. 9602, 3109601, Haifa, Israel.
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Tala Palchan-Hazan
- Department of Nuclear Medicine, Rambam Health Care Campus, P.O.B. 9602, 3109601, Haifa, Israel
| | - Quentin Maronnier
- Medical Imaging Department, Oncopole Claudius Regaud, Toulouse, France
| | - Olivier Caselles
- Medical Imaging Department, Oncopole Claudius Regaud, Toulouse, France
| | - Frédéric Courbon
- Medical Imaging Department, Oncopole Claudius Regaud, Toulouse, France
| | - Moshe Levy
- GE Healthcare, Tirat HaCarmel, Tirat HaCarmel, Israel
| | - Zohar Keidar
- Department of Nuclear Medicine, Rambam Health Care Campus, P.O.B. 9602, 3109601, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
13
|
Tvilum M, Knap MM, Hoffmann L, Khalil AA, Appelt AL, Haraldsen A, Alber M, Grau C, Schmidt HH, Kandi M, Holt MI, Lutz CM, Møller DS. Early radiologic and metabolic tumour response assessment during combined chemo-radiotherapy for locally advanced NSCLC. Clin Transl Radiat Oncol 2024; 45:100737. [PMID: 38317680 PMCID: PMC10839576 DOI: 10.1016/j.ctro.2024.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Background The role of early treatment response for patients with locally advanced non-small cell lung cancer (LA-NSCLC) treated with concurrent chemo-radiotherapy (cCRT) is unclear. The study aims to investigate the predictive value of response to induction chemotherapy (iCX) and the correlation with pattern of failure (PoF). Materials and methods Patients with LA-NSCLC treated with cCRT were included for analyses (n = 276). Target delineations were registered from radiotherapy planning PET/CT to diagnostic PET/CT, in between which patients received iCX. Volume, sphericity, and SUVpeak were extracted from each scan. First site of failure was categorised as loco-regional (LR), distant (DM), or simultaneous LR+M (LR+M). Fine and Gray models for PoF were performed: a baseline model (including performance status (PS), stage, and histology), an image model for squamous cell carcinoma (SCC), and an image model for non-SCC. Parameters included PS, volume (VOL) of tumour, VOL of lymph nodes, ΔVOL, sphericity, SUVpeak, ΔSUVpeak, and oligometastatic disease. Results Median follow-up was 7.6 years. SCC had higher sub-distribution hazard ratio (sHR) for LRF (sHR = 2.771 [1.577:4.87], p < 0.01) and decreased sHR for DM (sHR = 0.247 [0.125:0.485], p < 0.01). For both image models, high diagnostic SUVpeak increased risk of LRF (sHR = 1.059 [1.05:1.106], p < 0.01 for SCC, sHR = 1.12 [1.03:1.21], p < 0.01 for non-SCC). Patients with SCC and less decrease in VOL had higher sHR for DM (sHR = 1.025[1.001:1.048] pr. % increase, p = 0.038). Conclusion Poor response in disease volume was correlated with higher sHR of DM for SCC, no other clear correlation of response and PoF was observed. Histology significantly correlated with PoF with SCC prone to LRF and non-SCC prone to DM as first site of failure. High SUVpeak at diagnosis increased the risk of LRF for both histologies.
Collapse
Affiliation(s)
- Marie Tvilum
- Department of Oncology, Aarhus University Hospital, Denmark
- Danish Center for Particle Therapy, Aarhus University Hospital, Denmark
| | | | - Lone Hoffmann
- Department of Oncology, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Faculty of Health Sciences, Aarhus University, Denmark
| | | | - Ane L. Appelt
- Leeds Institute of Medical Research at St James’s, University of Leeds, United Kingdom
- Leeds Cancer Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Ate Haraldsen
- Department of Nuclear Medicine and PET-centre, Aarhus University Hospital, Denmark
| | - Markus Alber
- Department of Radiation Oncology, Heidelberg University Hospital, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg University Hospital, Germany
| | - Cai Grau
- Danish Center for Particle Therapy, Aarhus University Hospital, Denmark
| | | | - Maria Kandi
- Department of Oncology, Aarhus University Hospital, Denmark
| | | | | | - Ditte Sloth Møller
- Department of Oncology, Aarhus University Hospital, Denmark
- Department of Clinical Medicine, Faculty of Health Sciences, Aarhus University, Denmark
| |
Collapse
|
14
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Nazerani-Zemann T, Pernthaler B, Schwantzer G, Gstettner C. The systemic impact of different COVID-19 vaccines in 2-[18F] FDG-PET/CT. Sci Rep 2023; 13:21838. [PMID: 38071353 PMCID: PMC10710488 DOI: 10.1038/s41598-023-49376-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Austria started its COVID-19-vaccination program in December 2020 with three different vaccines. As the vaccination program continues, we encountered increased 2-[18F] FDG-activity not only in axillary lymph nodes ipsilateral to the injection site but also in other organs. The aim of this retrospective study is to present results of the metabolic activity of ipsilateral axillary lymph nodes, liver, blood pool, spleen, and bone marrow after three different vaccines. To our knowledge, this is the first study to examine systemic response changes in relation to time after COVID-19 vaccination using three different vaccines. The collected data of 220 eligible vaccinated patients (127 with BioNTech/Pfizer BNT162b2, 61 with Moderna, and 32 with AstraZeneca) examined with 2-[18F] FDG-PET/CT were enrolled. The PET/CT examinations were evaluated from day 1 to day 135 (SD: 23.2, median: 26) after different vaccinations. Seventy-one out of these 220 patients underwent a pre-vaccination 2-[18F] FDG -PET/CT. SUVmax of axillary node(s), and blood pool, liver, spleen, and bone marrow as reference organs were calculated. The ratio of SUVmax activity of axillary lymph node to reference organs was also compared in all patients. The tracer activity dynamics were investigated in three different vaccines. After BioNTech/Pfizer vaccination 2-[18F] FDG activity in axillary lymph nodes shows a steady decrease in all patients. Ten days after vaccination the 2-[18F] FDG uptake was at its highest activity. Seventy days after vaccination, tracer activity is not different from the background activity of 2-[18F] FDG in the axillary region. This result also applies to other two vaccines; however, in the 4th week after Moderna vaccination SUVmax in lymph nodes showed the highest peak of tracer activity. With AstraZeneca the highest activity was at the earlier days. There was no significant statistical difference of SUVmax of lymph nodes or its ratios to other reference organs between three groups of vaccines. SUVmax in lymph nodes was statistically significant lower than SUVmax in the liver, spleen, and bone marrow with p-values of < 0.001, 0.044, and 0.001, respectively. In the group of 71 patients with a pre-vaccination PET/CT examination, the median SUVmax of lymph nodes increased significantly after vaccination from 0.82 (IQR 0.59-1.38) to 1.80 (IQR 1.07-3.89)(p < 0.001). In contrast median tracer activity in the liver decreased from 3.37 (IQR 2.83-3.91) to 3.11 (2.56-3.70) (p = 0.032). There was no significant change of tracer activity after vaccination in other reference regions (mediastinum, spleen, and bone marrow). In this group of 71 patients, there was also no significant difference in tracer activity in different types of vaccines. Local site and ipsilateral axillary lymph node activity in 2-[18F] FDG PET/CT after COVID19-vaccination is suggested in many studies. The main challenge is recognizing the changes in lymph nodes during time after vaccination to minimize false interpretation, foremost in patients with oncological diagnoses. Moreover, different vaccines cause different system metabolic changes. The knowledge of vaccine type, the time interval between vaccination and PET/CT scan is essential, especially in therapy evaluation.
Collapse
Affiliation(s)
- Tina Nazerani-Zemann
- Division of Nuclear Medicine, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9A, 8038, Graz, Austria.
| | - Birgit Pernthaler
- Division of Nuclear Medicine, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9A, 8038, Graz, Austria
| | - Gerold Schwantzer
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Christian Gstettner
- Division of Nuclear Medicine, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9A, 8038, Graz, Austria
| |
Collapse
|
16
|
Zirakchian Zadeh M. Clinical Application of 18F-FDG-PET Quantification in Hematological Malignancies: Emphasizing Multiple Myeloma, Lymphoma and Chronic Lymphocytic Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:800-814. [PMID: 37558532 DOI: 10.1016/j.clml.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
Most hematological malignancies display heightened glycolytic activity, leading to their detectability through 18F-FDG-PET imaging. PET quantification enables the extraction of metabolic information from tumors. Among various PET measurements, maximum standardized uptake value (SUVmax), which indicates the highest value of 18F-FDG uptake within the tumor, has emerged as the commonly used parameter in clinical oncology. This is because of SUVmax ease of calculation using most available commercial workstations, as well as its simplicity and independence from observer interpretation. Nonetheless, SUVmax represents the increase in activity within a specific small area, which may not fully capture the overall tumor uptake. Volumetric PET parameters have been identified as a potential solution to overcome certain limitations associated with SUVmax. However, these parameters are influenced by the low spatial resolution of PET when assessing small lesions. Another challenge is the high number of lesions observed in some patients, leading to a time-consuming process for evaluating all focal lesions. Some institutions recently have started advocating for CT-based segmentation as a method for measuring radiotracer uptake in the bone marrow and overall bone of the patients. This review article aims to provide insights into clinical application of PET quantification specifically focusing on 3 major hematologic malignancies: multiple myeloma, lymphoma, and chronic lymphocytic leukemia.
Collapse
Affiliation(s)
- Mahdi Zirakchian Zadeh
- Molecular Imaging and Therapy and Interventional Radiology Services, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
17
|
Mohr P, van Sluis J, Providência L, van Snick JH, Lub-de Hooge MN, Willemsen AT, Glaudemans AWJM, Boellaard R, Lammertsma AA, Brouwers AH, Tsoumpas C. Long Versus Short Axial Field of View Immuno-PET/CT: Semiquantitative Evaluation for 89Zr-Trastuzumab. J Nucl Med 2023; 64:1815-1820. [PMID: 37536740 DOI: 10.2967/jnumed.123.265621] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/20/2023] [Indexed: 08/05/2023] Open
Abstract
The purpose of this study was to quantify any differences between the SUVs of 89Zr immuno-PET scans obtained using a PET/CT system with a long axial field of view (LAFOV; Biograph Vision Quadra) compared to a PET/CT system with a short axial field of view (SAFOV; Biograph Vision) and to evaluate how LAFOV PET scan duration affects image noise and SUV metrics. Methods: Five metastatic breast cancer patients were scanned consecutively on SAFOV and LAFOV PET/CT scanners. Four additional patients were scanned using only LAFOV PET/CT. Scans on both systems lasted approximately 30 min and were acquired 4 d after injection of 37 MBq of 89Zr-trastuzumab. LAFOV list-mode data were reprocessed to obtain images acquired using shorter scan durations (15, 10, 7.5, 5, and 3 min). Volumes of interest were placed in healthy tissues, and tumors were segmented semiautomatically to compare coefficients of variation and to perform Bland-Altman analysis on SUV metrics (SUVmax, SUVpeak, and SUVmean). Results: Using 30-min images, 2 commonly used lesion SUV metrics were higher for SAFOV than for LAFOV PET (SUVmax, 16.2% ± 13.4%, and SUVpeak, 10.1% ± 7.2%), whereas the SUVmean of healthy tissues showed minimal differences (0.7% ± 5.8%). Coefficients of variation in the liver derived from 30-min SAFOV PET were between those of 3- and 5-min LAFOV PET. The smallest SUVmax and SUVpeak differences between SAFOV and LAFOV were found for 3-min LAFOV PET. Conclusion: LAFOV 89Zr immuno-PET showed a lower SUVmax and SUVpeak than SAFOV because of lower image noise. LAFOV PET scan duration may be reduced at the expense of increasing image noise and bias in SUV metrics. Nevertheless, SUVpeak showed only minimal bias when reducing scan duration from 30 to 10 min.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Providência
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johannes H van Snick
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Antoon T Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
Schmidt FP, Mannheim JG, Linder PM, Will P, Kiefer LS, Conti M, la Fougère C, Rausch I. Impact of the maximum ring difference on image quality and noise characteristics of a total-body PET/CT scanner. Z Med Phys 2023:S0939-3889(23)00113-7. [PMID: 37867050 DOI: 10.1016/j.zemedi.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/14/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023]
Abstract
The sensitivity of a PET system highly depends on the axial acceptance angle or maximum ring difference (MRD), which can be particularly high for total-body scanners due to their larger axial field of views (aFOVs). This study aims to evaluate the impact on image quality (IQ) and noise performance when MRD85 (18°), the current standard for clinical use, is increased to MRD322 (52°) for the Biograph Vision Quadra (Siemens Healthineers). METHODS Studies with a cylindrical phantom covering the 106 cm aFOV and an IEC phantom filled with 18F, 68Ga and 89Zr were performed for acquisition times from 60 to 1800 s and activity concentrations from 0.4 to 3 kBq/ml to assess uniformity, contrast recovery coefficients (CRCs) and to characterize noise by coefficient of variation (CV). Spatial resolution was compared for both MRDs by sampling a quadrant of the FOV with a point source. Further IQ, CV, liver SUVmean and SUVmax were compared for a cohort of 5 patients scanned with [18F]FDG (3 MBq/kg, 1 h p.i.) from 30 to 300 s. RESULTS CV was improved by a factor of up to 1.49 and is highest for short acquisition times, peaks at the center field of view and mitigates parabolic in axial direction with no difference to MRD85 beyond the central 80 cm. No substantial differences between the two evaluated MRDs in regards to uniformity, SUVmean or CRC for the different isotopes were observed. A degradation of the average spatial resolution of 0.9 ± 0.2 mm in the central 40 cm FOV was determined with MRD322. Depending on the acquisition time MRD322 resulted in a decrease of SUVmax between 23.8% (30 s) and 9.0% (300 s). CONCLUSION Patient and phantom studies revealed that scan time could be lowered by approximately a factor of two with MRD322 while maintaining similar noise performance. The moderate degradation in spatial resolution for MRD322 is worth to exploit the full potential of the Quadra by either shorten scan times or leverage noise performance in particular for low count scenarios such as ultra-late imaging or dynamic studies with high temporal resolution.
Collapse
Affiliation(s)
- F P Schmidt
- Department of Nuclear Medicine and Clinical Molecular Imaging, University hospital Tuebingen, Tuebingen, Germany; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, Tuebingen, Germany.
| | - J G Mannheim
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - P M Linder
- Department of Nuclear Medicine and Clinical Molecular Imaging, University hospital Tuebingen, Tuebingen, Germany
| | - P Will
- Department of Nuclear Medicine and Clinical Molecular Imaging, University hospital Tuebingen, Tuebingen, Germany
| | - L S Kiefer
- Department of Nuclear Medicine and Clinical Molecular Imaging, University hospital Tuebingen, Tuebingen, Germany
| | - M Conti
- Siemens Medical Solutions USA Inc., Molecular Imaging, Knoxville, TN, USA
| | - C la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, University hospital Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - I Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Calderón E, Schmidt FP, Lan W, Castaneda-Vega S, Brendlin AS, Trautwein NF, Dittmann H, la Fougère C, Kiefer LS. Image Quality and Quantitative PET Parameters of Low-Dose [ 18F]FDG PET in a Long Axial Field-of-View PET/CT Scanner. Diagnostics (Basel) 2023; 13:3240. [PMID: 37892061 PMCID: PMC10606613 DOI: 10.3390/diagnostics13203240] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/15/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
PET/CT scanners with a long axial field-of-view (LAFOV) provide increased sensitivity, enabling the adjustment of imaging parameters by reducing the injected activity or shortening the acquisition time. This study aimed to evaluate the limitations of reduced [18F]FDG activity doses on image quality, lesion detectability, and the quantification of lesion uptake in the Biograph Vision Quadra, as well as to assess the benefits of the recently introduced ultra-high sensitivity mode in a clinical setting. A number of 26 patients who underwent [18F]FDG-PET/CT (3.0 MBq/kg, 5 min scan time) were included in this analysis. The PET raw data was rebinned for shorter frame durations to simulate 5 min scans with lower activities in the high sensitivity (HS) and ultra-high sensitivity (UHS) modes. Image quality, noise, and lesion detectability (n = 82) were assessed using a 5-point Likert scale. The coefficient of variation (CoV), signal-to-noise ratio (SNR), tumor-to-background ratio (TBR), and standardized uptake values (SUV) including SUVmean, SUVmax, and SUVpeak were evaluated. Subjective image ratings were generally superior in UHS compared to the HS mode. At 0.5 MBq/kg, lesion detectability decreased to 95% (HS) and to 98% (UHS). SNR was comparable at 1.0 MBq/kg in HS (5.7 ± 0.6) and 0.5 MBq/kg in UHS (5.5 ± 0.5). With lower doses, there were negligible reductions in SUVmean and SUVpeak, whereas SUVmax increased steadily. Reducing the [18F]FDG activity to 1.0 MBq/kg (HS/UHS) in a LAFOV PET/CT provides diagnostic image quality without statistically significant changes in the uptake parameters. The UHS mode improves image quality, noise, and lesion detectability compared to the HS mode.
Collapse
Affiliation(s)
- Eduardo Calderón
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
| | - Fabian P. Schmidt
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Wenhong Lan
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
| | - Salvador Castaneda-Vega
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Andreas S. Brendlin
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, 72076 Tuebingen, Germany;
| | - Nils F. Trautwein
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
| | - Christian la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72074 Tuebingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tuebingen, 72074 Tuebingen, Germany
| | - Lena Sophie Kiefer
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, 72076 Tuebingen, Germany; (E.C.); (F.P.S.); (W.L.); (S.C.-V.); (N.F.T.); (H.D.); (C.l.F.)
| |
Collapse
|
20
|
Kendrick J, Francis RJ, Hassan GM, Rowshanfarzad P, Ong JSL, Barry N, Rusanov B, Ebert MA. Quantitative [ 68Ga]Ga-PSMA-11 PET biomarkers for the analysis of lesion-level progression in biochemically recurrent prostate cancer: a multicentre study. Sci Rep 2023; 13:17673. [PMID: 37848692 PMCID: PMC10582101 DOI: 10.1038/s41598-023-45106-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023] Open
Abstract
[68Ga]Ga-PSMA-11 PET has become the standard imaging modality for biochemically recurrent (BCR) prostate cancer (PCa). However, its prognostic value in assessing response at this stage remains uncertain. The study aimed to assess the prognostic significance of radiographic patient-level patterns of progression derived from lesion-level biomarker quantitation in metastatic disease sites. A total of 138 BCR PCa patients with both baseline and follow-up [68Ga]Ga-PSMA-11 PET scans were included in this analysis. Tumour response was quantified at the lesion level using commonly used quantitative parameters (SUVmean, SUVmax, SUVpeak, volume), and patients were classified as systemic, mixed, or no-progression based on these response classifications. A total of 328 matched lesions between baseline and follow-up scans were analysed. The results showed that systemic progressors had a significantly higher risk of death than patients with no progression with SUVmean demonstrating the highest prognostic value (HR = 5.70, 95% CI = 2.63-12.37, p < 0.001, C-Index = 0.69). Moreover, progressive disease as measured by SUVmean using the radiographic PSMA PET Progression Criteria (rPPP) was found to be significantly prognostic for patient overall survival (HR = 3.67, 95% CI = 1.82-7.39, p < 0.001, C-Index = 0.65). This work provides important evidence supporting the prognostic utility of PSMA response quantitation in the BCR setting.
Collapse
Affiliation(s)
- Jake Kendrick
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia.
- Centre for Advanced Technologies in Cancer Research, Perth, WA, Australia.
| | - Roslyn J Francis
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Australian Centre for Quantitative Imaging, University of Western Australia, Crawley, WA, Australia
| | - Ghulam Mubashar Hassan
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia
- Centre for Advanced Technologies in Cancer Research, Perth, WA, Australia
| | - Jeremy S L Ong
- Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Nathaniel Barry
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia
- Centre for Advanced Technologies in Cancer Research, Perth, WA, Australia
| | - Branimir Rusanov
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia
- Centre for Advanced Technologies in Cancer Research, Perth, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Martin A Ebert
- School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA, Australia
- Centre for Advanced Technologies in Cancer Research, Perth, WA, Australia
- Australian Centre for Quantitative Imaging, University of Western Australia, Crawley, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- 5D Clinics, Claremont, WA, Australia
| |
Collapse
|
21
|
Mokoala KMG, Lawal IO, Maserumule LC, Bida M, Maes A, Ndlovu H, Reed J, Mahapane J, Davis C, Van de Wiele C, Popoola G, Giesel FL, Vorster M, Sathekge MM. Correlation between [ 68Ga]Ga-FAPI-46 PET Imaging and HIF-1α Immunohistochemical Analysis in Cervical Cancer: Proof-of-Concept. Cancers (Basel) 2023; 15:3953. [PMID: 37568769 PMCID: PMC10417683 DOI: 10.3390/cancers15153953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Hypoxia leads to changes in tumor microenvironment (upregulated CAFs) with resultant aggressiveness. A key factor in the physiological response to hypoxia is hypoxia-inducible factor-1alpha (HIF-1α). [68Ga]Ga-FAPI PET imaging has been demonstrated in various cancer types. We hypothesized that [68Ga]Ga-FAPI PET may be used as an indirect tracer for mapping hypoxia by correlating the image findings to pathological analysis of HIF-1α expression. The [68Ga]Ga-FAPI PET/CT scans of women with cancer of the cervix were reviewed and the maximum and mean standardized uptake value (SUVmax and SUVmean) and FAPI tumor volume (FAPI-TV) were documented. Correlation analysis was performed between PET-derived parameters and immunohistochemical staining as well as between PET-derived parameters and the presence of metastasis. Ten women were included. All patients demonstrated tracer uptake in the primary site or region of the primary. All patients had lymph node metastases while only six patients had distant visceral or skeletal metastases. The mean SUVmax, SUVmean, and FAPI-TV was 18.89, 6.88, and 195.66 cm3, respectively. The average FAPI-TV for patients with additional sites of metastases was higher than those without. Immunohistochemistry revealed varying intensities of HIF-1α expression in all tested samples. There was a positive correlation between the presence of skeletal metastases and staining for HIF-1α (r=0.80;p=0.017). The presence of skeletal metastasis was correlated to the HIF-1⍺ staining (percentage distribution). Furthermore, the FAPI-TV was a better predictor of metastatic disease than the SUVmax.
Collapse
Affiliation(s)
- Kgomotso M. G. Mokoala
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
| | - Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa
| | - Letjie C. Maserumule
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
| | - Meshack Bida
- National Health Laboratory Services, Department of Anatomical Pathology, Pretoria 0001, South Africa;
| | - Alex Maes
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
- Katholieke University Leuven, 3000 Kortrijk, Belgium
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
| | - Janet Reed
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
| | - Johncy Mahapane
- Department of Radiography, University of Pretoria, Pretoria 0028, South Africa;
| | - Cindy Davis
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
| | - Christophe Van de Wiele
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
- Department of Diagnostic Sciences, University Ghent, 9000 Ghent, Belgium
| | - Gbenga Popoola
- Lincolnshire Partnership NHS Foundation Trust, St George’s, Lincoln, Lincolnshire LN1 1FS, UK;
| | - Frederik L. Giesel
- Department of Nuclear Medicine, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Mariza Vorster
- Department of Nuclear Medicine, University of Kwazulu Natal, Durban 4001, South Africa;
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0028, South Africa; (K.M.G.M.); (I.O.L.); (L.C.M.); (A.M.); (H.N.); (J.R.); (C.D.); (C.V.d.W.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa
| |
Collapse
|
22
|
Galvez T, Berkane I, Thézenas S, Eberlé MC, Flori N, Guillemard S, Ilonca AD, Santoro L, Kotzki PO, Senesse P, Deshayes E. Identification of 18F-FDG PET/CT Parameters Associated with Weight Loss in Patients with Esophageal Cancer. Nutrients 2023; 15:3042. [PMID: 37447367 DOI: 10.3390/nu15133042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/16/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
18F-FDG PET-CT is routinely performed as part of the initial staging of numerous cancers. Other than having descriptive, predictive and prognostic values for tumors, 18F-FDG PET-CT provides full-body data, which could inform on concurrent pathophysiological processes such as malnutrition. To test this hypothesis, we measured the 18F-FDG uptake in several organs and evaluated their association with weight loss in patients at diagnosis of esophageal cancer. Forty-eight patients were included in this retrospective monocentric study. 18F-FDG uptake quantification was performed in the brain, the liver, the spleen, bone marrow, muscle and the esophageal tumor itself and was compared between patients with different amounts of weight loss. We found that Total Lesion Glycolysis (TLG) and peak Standardized Uptake Values (SUVpeak) measured in the brain correlated with the amount of weight loss: TLG was, on average, higher in patients who had lost more than 5% of their usual weight, whereas brain SUVpeak were, on average, lower in patients who had lost more than 10% of their weight. Higher TLG and lower brain SUVpeak were associated with worse OS in the univariate analysis. This study reports a new and significant association between 18F-FDG uptake in the brain and initial weight loss in patients with esophageal cancer.
Collapse
Affiliation(s)
- Thierry Galvez
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Department of Endocrinology, Diabetes and Nutrition, CHU de Montpellier, Université de Montpellier, 34295 Montpellier, France
| | - Ikrame Berkane
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Simon Thézenas
- Biometry Unit, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Marie-Claude Eberlé
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Nicolas Flori
- Department of Clinical Nutrition and Gastroenterology, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Sophie Guillemard
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Alina Diana Ilonca
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Lore Santoro
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
| | - Pierre-Olivier Kotzki
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Pierre Senesse
- Department of Clinical Nutrition and Gastroenterology, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| | - Emmanuel Deshayes
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, 34298 Montpellier, Cedex 5, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, 34298 Montpellier, France
| |
Collapse
|
23
|
van Sluis J, Boellaard R, Dierckx RAJO, van Esch ELM, Croes DA, de Ruijter LK, van de Donk PP, de Vries EGE, Noordzij W, Brouwers AH. Optimisation of scan duration and image quality in oncological 89Zr immunoPET imaging using the Biograph Vision PET/CT. Eur J Nucl Med Mol Imaging 2023; 50:2258-2270. [PMID: 36947185 PMCID: PMC10250429 DOI: 10.1007/s00259-023-06194-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/07/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE Monoclonal antibody (mAb)-based PET (immunoPET) imaging can characterise tumour lesions non-invasively. It may be a valuable tool to determine which patients may benefit from treatment with a specific monoclonal antibody (mAb) and evaluate treatment response. For 89Zr immunoPET imaging, higher sensitivity of state-of-the art PET/CT systems equipped with silicon photomultiplier (SiPM)-based detector elements may be beneficial as the low positron abundance of 89Zr causes a low signal-to-noise level. Moreover, the long physical half-life limits the amount of activity that can be administered to the patients leading to poor image quality even when using long scan durations. Here, we investigated the difference in semiquantitative performance between the PMT-based Biograph mCT, our clinical reference system, and the SiPM-based Biograph Vision PET/CT in 89Zr immunoPET imaging. Furthermore, the effects of scan duration reduction using the Vision on semiquantitative imaging parameters and its influence on image quality assessment were evaluated. METHODS Data were acquired on day 4 post 37 MBq 89Zr-labelled mAb injection. Five patients underwent a double scan protocol on both systems. Ten patients were scanned only on the Vision. For PET image reconstruction, three protocols were used, i.e. one camera-dependent protocol and European Association of Nuclear Medicine Research Limited (EARL) standards 1 and 2 compliant protocols. Vision data were acquired in listmode and were reprocessed to obtain images at shorter scan durations. Semiquantitative PET image parameters were derived from tumour lesions and healthy tissues to assess differences between systems and scan durations. Differently reconstructed images obtained using the Vision were visually scored regarding image quality by two nuclear medicine physicians. RESULTS When images were reconstructed using 100% acquisition time on both systems following EARL standard 1 compliant reconstruction protocols, results regarding semiquantification were comparable. For Vision data, reconstructed images that conform to EARL1 standards still resulted in comparable semiquantification at shorter scan durations (75% and 50%) regarding 100% acquisition time. CONCLUSION Scan duration of 89Zr immunoPET imaging using the Vision can be decreased up to 50% compared with using the mCT while maintaining image quality using the EARL1 compliant reconstruction protocol.
Collapse
Affiliation(s)
- Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Centers Amsterdam, Free University of Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Evelien L M van Esch
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Demi A Croes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Laura Kist de Ruijter
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Pim P van de Donk
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
24
|
Wang Y, Chow D, Indrakanti SS, Palmer EL, Scott JA. Schrodinger's cat and Deauville 5 point scoring. Clin Imaging 2023; 101:180-182. [PMID: 37385118 DOI: 10.1016/j.clinimag.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 06/08/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Affiliation(s)
- Yingbing Wang
- Massachusetts General Hospital, Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, 55 Fruit Street, White 4-427, Boston, MA 02114, United States of America.
| | - David Chow
- Massachusetts General Hospital, Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, 55 Fruit Street, White 4-427, Boston, MA 02114, United States of America
| | - Shalini Santoshi Indrakanti
- Massachusetts General Hospital, Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, 55 Fruit Street, White 4-427, Boston, MA 02114, United States of America
| | - Edwin Lincoln Palmer
- Massachusetts General Hospital, Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, 55 Fruit Street, White 4-427, Boston, MA 02114, United States of America
| | - James Arthur Scott
- Massachusetts General Hospital, Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, 55 Fruit Street, White 4-427, Boston, MA 02114, United States of America
| |
Collapse
|
25
|
Sipilä O, Liukkonen J, Halme HL, Tolvanen T, Sohlberg A, Hakulinen M, Manninen AL, Tahvanainen K, Tunninen V, Ollikainen T, Kangasmaa T, Kangasmäki A, Vuorela J. Variability in PET image quality and quantification measured with a permanently filled 68Ge-phantom: a multi-center study. EJNMMI Phys 2023; 10:38. [PMID: 37322376 DOI: 10.1186/s40658-023-00551-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND This study evaluated, as a snapshot, the variability in quantification and image quality (IQ) of the clinically utilized PET [18F]FDG whole-body protocols in Finland using a NEMA/IEC IQ phantom permanently filled with 68Ge. METHODS The phantom was imaged on 14 PET-CT scanners, including a variety of models from two major vendors. The variability of the recovery coefficients (RCmax, RCmean and RCpeak) of the hot spheres as well as percent background variability (PBV), coefficient of variation of the background (COVBG) and accuracy of corrections (AOC) were studied using images from clinical and standardized protocols with 20 repeated measurements. The ranges of the RCs were also compared to the limits of the EARL 18F standards 2 accreditation (EARL2). The impact of image noise on these parameters was studied using averaged images (AVIs). RESULTS The largest variability in RC values of the routine protocols was found for the RCmax with a range of 68% and with 10% intra-scanner variability, decreasing to 36% when excluding protocols with suspected cross-calibration failure or without point-spread-function (PSF) correction. The RC ranges of individual hot spheres in routine or standardized protocols or AVIs fulfilled the EARL2 ranges with two minor exceptions, but fulfilling the exact EARL2 limits for all hot spheres was variable. RCpeak was less dependent on averaging and reconstruction parameters than RCmax and RCmean. The PBV, COVBG and AOC varied between 2.3-11.8%, 9.6-17.8% and 4.8-32.0%, respectively, for the routine protocols. The RC ranges, PBV and COVBG were decreased when using AVIs. With AOC, when excluding routine protocols without PSF correction, the maximum value dropped to 15.5%. CONCLUSION The maximum variability of the RC values for the [18F]FDG whole-body protocols was about 60%. The RC ranges of properly cross-calibrated scanners with PSF correction fitted to the EARL2 RC ranges for individual sphere sizes, but fulfilling the exact RC limits would have needed further optimization. RCpeak was the most robust RC measure. Besides COVBG, also RCs and PVB were sensitive to image noise.
Collapse
Affiliation(s)
- O Sipilä
- HUS Diagnostic Center, Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, P. O. Box 442, 00029, Helsinki, Finland.
| | - J Liukkonen
- Radiation and Nuclear Safety Authority, Vantaa, Finland
| | - H-L Halme
- HUS Diagnostic Center, Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, P. O. Box 442, 00029, Helsinki, Finland
| | - T Tolvanen
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - A Sohlberg
- Department of Nuclear Medicine, Päijät-Häme Central Hospital, Lahti, Finland
| | - M Hakulinen
- Department of Clinical Physiology and Nuclear Medicine, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - A-L Manninen
- OYS Department of Nuclear Medicine and Radiology, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - K Tahvanainen
- HUS Diagnostic Center, Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, P. O. Box 442, 00029, Helsinki, Finland
| | - V Tunninen
- Department of Clinical Physiology and Nuclear Medicine, Satakunta Central Hospital, Pori, Finland
| | - T Ollikainen
- Clinical Physiology and Neurophysiology, North Karelia Central Hospital, Joensuu, Finland
| | - T Kangasmaa
- Department of Clinical Physiology and Nuclear Medicine, Vaasa Central Hospital, Wellbeing Services County of Ostrobothnia, Vaasa, Finland
| | - A Kangasmäki
- Department of Imaging and Radiotherapy, Docrates Cancer Center, Helsinki, Finland
| | - J Vuorela
- Clinical Physiology and Nuclear Medicine, Central Finland Health Care District, Jyväskylä, Finland
| |
Collapse
|
26
|
Rajagopal A, Natsuaki Y, Wangerin K, Hamdi M, An H, Sunderland JJ, Laforest R, Kinahan PE, Larson PEZ, Hope TA. Synthetic PET via Domain Translation of 3-D MRI. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2023; 7:333-343. [PMID: 37396797 PMCID: PMC10311993 DOI: 10.1109/trpms.2022.3223275] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Historically, patient datasets have been used to develop and validate various reconstruction algorithms for PET/MRI and PET/CT. To enable such algorithm development, without the need for acquiring hundreds of patient exams, in this article we demonstrate a deep learning technique to generate synthetic but realistic whole-body PET sinograms from abundantly available whole-body MRI. Specifically, we use a dataset of 56 18F-FDG-PET/MRI exams to train a 3-D residual UNet to predict physiologic PET uptake from whole-body T1-weighted MRI. In training, we implemented a balanced loss function to generate realistic uptake across a large dynamic range and computed losses along tomographic lines of response to mimic the PET acquisition. The predicted PET images are forward projected to produce synthetic PET (sPET) time-of-flight (ToF) sinograms that can be used with vendor-provided PET reconstruction algorithms, including using CT-based attenuation correction (CTAC) and MR-based attenuation correction (MRAC). The resulting synthetic data recapitulates physiologic 18F-FDG uptake, e.g., high uptake localized to the brain and bladder, as well as uptake in liver, kidneys, heart, and muscle. To simulate abnormalities with high uptake, we also insert synthetic lesions. We demonstrate that this sPET data can be used interchangeably with real PET data for the PET quantification task of comparing CTAC and MRAC methods, achieving ≤ 7.6% error in mean-SUV compared to using real data. These results together show that the proposed sPET data pipeline can be reasonably used for development, evaluation, and validation of PET/MRI reconstruction methods.
Collapse
Affiliation(s)
- Abhejit Rajagopal
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA 94158 USA
| | - Yutaka Natsuaki
- Department of Radiation Oncology, University of New Mexico, Albuquerque, NM 87131 USA
| | | | - Mahdjoub Hamdi
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63130 USA
| | - Hongyu An
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63130 USA
| | - John J Sunderland
- Department of Radiology, The University of Iowa, Iowa City, IA 52242 USA
| | - Richard Laforest
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63130 USA
| | - Paul E Kinahan
- Department of Radiology, University of Washington, Seattle, WA 98195 USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA 94158 USA
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, CA 94158 USA
| |
Collapse
|
27
|
Ahlman MA, Grayson PC. Advanced molecular imaging in large-vessel vasculitis: Adopting FDG-PET into a clinical workflow. Best Pract Res Clin Rheumatol 2023; 37:101856. [PMID: 37516606 PMCID: PMC10818007 DOI: 10.1016/j.berh.2023.101856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023]
Abstract
The use of fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging to detect vascular inflammation is increasingly common in the clinical management of patients with large-vessel vasculitis (LVV). In this review, the role of FDG-PET imaging to diagnose and monitor vascular disease activity will be detailed. Suggestions on incorporation of FDG-PET imaging into a clinical workflow will be provided with emphasis on patient preparation, image acquisition, and image interpretation. If FDG-PET imaging is obtained, multimodal imaging assessment, whereby FDG-PET imaging and non-invasive angiography are obtained concurrently, and correlation of imaging findings with clinical assessment is generally advisable. Considering the clinical scenario and treatment status of the patient is important when interpreting vascular FDG-PET image findings.
Collapse
Affiliation(s)
- Mark A Ahlman
- Nuclear Medicine Department, Medical College of Georgia, Augusta, GA, USA
| | - Peter C Grayson
- National Institutes of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, USA.
| |
Collapse
|
28
|
Li Y, Sijtsema NM, de Vette SPM, Steenbakkers RJHM, Zhang F, Noordzij W, Van den Bosch L, Langendijk JA, van Dijk LV. Validation of the 18F-FDG PET image biomarker model predicting late xerostomia after head and neck cancer radiotherapy. Radiother Oncol 2023; 180:109458. [PMID: 36608769 DOI: 10.1016/j.radonc.2022.109458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Previously, PET image biomarkers (PET-IBMs) - the 90th percentile standardized uptake value (P90-SUV) and the Mean SUV (Mean-SUV) of the contralateral parotid gland (cPG) - were identified as predictors for late-xerostomia following head and neck cancer (HNC) radiotherapy. The aim of the current study was to assess in an independent validation cohort whether these pre-treatment PET-IBM can improve late-xerostomia prediction compared to the prediction with baseline xerostomia and mean cPG dose alone. MATERIALS AND METHODS The prediction endpoint was patient-rated moderate-to-severe xerostomia at 12 months after radiotherapy. The PET-IBMs were extracted from pre-treatment 18 F-FDG PET images. The performance of the model (base model) with baseline xerostomia and mean cPG dose alone and models with additionally P90-SUV or Mean-SUV were tested in the current independent validation cohort. Specifically, model discrimination (area under the curve: AUC) and calibration (calibration plot) were evaluated. RESULTS The current validation cohort consisted of 137 patients of which 40% developed moderate-to-severe xerostomia at 12 months. Both the PET-P90 model (AUC:PET-P90 = 0.71) and the PET-Mean model (AUC: PET-Mean = 0.70) performed well in the current validation cohort. Moreover, their performance were improved compared to the base model (AUC:base model= 0.68). The calibration plots showed a good fit of the prediction to the actual rates for all tested models. CONCLUSION PET-IBMs showed an improved prediction of late-xerostomia when added to the base model in this validation cohort. This contributed to the published hypothesis that PET-IBMs include individualized information and can serve as a pre-treatment risk factor for late-xerostomia.
Collapse
Affiliation(s)
- Yan Li
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Nanna Maria Sijtsema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - Fan Zhang
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lisa Van den Bosch
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Johannes Albertus Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lisanne Vania van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
29
|
Yuge S, Miyake KK, Ishimori T, Kataoka M, Matsumoto Y, Fujimoto K, Sugie T, Toi M, Nakamoto Y. Reproducibility assessment of uptake on dedicated breast PET for noise discrimination. Ann Nucl Med 2023; 37:121-130. [PMID: 36434200 DOI: 10.1007/s12149-022-01809-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Dedicated breast PET (dbPET) systems have improved the detection of small breast cancers but have increased false-positive diagnoses due to an increased chance of noise detection. This study examined whether reproducibility assessment using paired images helped to improve noise discrimination and diagnostic performance in dbPET. METHODS This study included 21 patients with newly diagnosed breast cancer who underwent [18F]FDG-dbPET and contrast-enhanced breast MRI. A 10-min dbPET data scan was acquired per breast, and two sets of reconstructed images were generated (named dbPET-1 and dbPET-2, respectively), each of which consisted of randomly allocated 5-min data from the 10-min data. Uptake spots higher than the background were indexed for the study with visual assessment. All indexed uptakes on dbPET-1 were evaluated using dbPET-2 for reproducibility. MRI findings based on the Breast Imaging-Reporting and Data System (BI-RADS) 2013 were used as the gold standard. Uptake spots that corresponded to BI-RADS 1 on MRI were considered noise, while those with BI-RADS 4b-6 were considered malignancies. The diagnostic performance of dbPET for malignancy was evaluated using four different criteria: any uptake on dbPET-1 regarded as positive (criterion A), a subjective visual assessment of dbPET-1 (criterion B), reproducibility assessment between dbPET-1 and dbPET-2 (criterion C), and a combination of B and C (criterion D). RESULTS A total of 213 indexed uptake spots were identified on dbPET-1, including 152, 15, 6, 6, and 34 lesions classified as BI-RADS MRI categories 1, 2, 4b, 4c, and 5, respectively. Overall, 31.9% of the index uptake values were reproducible. All malignant lesions were reproducible, whereas 93.4% of noise was not reproducible. The sensitivities for malignancy for criteria A, B, C, and D were 100%, 91.3%, 100%, and 91.3%, respectively, with positive predictive values (PPVs) of 21.4%, 68.9%, 67.6%, and 82.4%, respectively. CONCLUSIONS Our results demonstrated that reproducibility assessment helped reduce false-positive findings caused by noise on dbPET without lowering the sensitivity for malignancy. While subjective visual assessment was also efficient in increasing PPV, it occasionally missed malignant uptake.
Collapse
Affiliation(s)
- Shunsuke Yuge
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| | - Kanae K Miyake
- Department of Advanced Medical Imaging Research, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto City, Kyoto, Japan, 606-8507.
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| | - Masako Kataoka
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| | - Yoshiaki Matsumoto
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| | - Koji Fujimoto
- Department of Real World Data Research and Development, Kyoto University Graduate School of Medicine, Kyoto City, Kyoto, Japan
| | - Tomoharu Sugie
- Department of Breast Surgery, Kansai Medical University Hospital, Hirakata City, Osaka, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto City, Kyoto, Japan
| |
Collapse
|
30
|
Shankar LK, Huang E, Litiere S, Hoekstra OS, Schwartz L, Collette S, Boellaard R, Bogaerts J, Seymour L, deVries EGE. Meta-Analysis of the Test-Retest Repeatability of [18F]-Fluorodeoxyglucose Standardized Uptake Values: Implications for Assessment of Tumor Response. Clin Cancer Res 2023; 29:143-153. [PMID: 36302172 DOI: 10.1158/1078-0432.ccr-21-3143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/28/2022] [Accepted: 10/24/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Currently, guidelines for PET with 18F-fluorodeoxyglucose (FDG-PET) interpretation for assessment of therapy response in oncology primarily involve visual evaluation of FDG-PET/CT scans. However, quantitative measurements of the metabolic activity in tumors may be even more useful in evaluating response to treatment. Guidelines based on such measurements, including the European Organization for Research and Treatment of Cancer Criteria and PET Response Criteria in Solid Tumors, have been proposed. However, more rigorous analysis of response criteria based on FDG-PET measurements is needed to adopt regular use in practice. EXPERIMENTAL DESIGN Well-defined boundaries of repeatability and reproducibility of quantitative measurements to discriminate noise from true signal changes are a needed initial step. An extension of the meta-analysis from de Langen and colleagues (2012) of the test-retest repeatability of quantitative FDG-PET measurements, including mean, maximum, and peak standardized uptake values (SUVmax, SUVmean, and SUVpeak, respectively), was performed. Data from 11 studies in the literature were used to estimate the relationship between the variance in test-retest measurements with uptake level and various study-level, patient-level, and lesion-level characteristics. RESULTS Test-retest repeatability of percentage fluctuations for all three types of SUV measurement (max, mean, and peak) improved with higher FDG uptake levels. Repeatability in all three SUV measurements varied for different lesion locations. Worse repeatability in SUVmean was also associated with higher tumor volumes. CONCLUSIONS On the basis of these results, recommendations regarding SUV measurements for assessing minimal detectable changes based on repeatability and reproducibility are proposed. These should be applied to differentiate between response categories for a future set of FDG-PET-based criteria that assess clinically significant changes in tumor response.
Collapse
Affiliation(s)
| | - Erich Huang
- National Cancer Institute, Bethesda, Maryland
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Otto S Hoekstra
- Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | - Larry Schwartz
- Columbia University Medical Center and New York Presbyterian Hospital, New York, New York
| | | | | | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | | |
Collapse
|
31
|
Itagaki K, Mitsumoto K, Kajisako M, Shioji M, Kawase S. Effect of Tumor-Pixel Positioning on the Variability of SUV Measurements in PET Images. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2023; 11:71-81. [PMID: 36619185 PMCID: PMC9803621 DOI: 10.22038/aojnmb.2022.61623.1434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/04/2022] [Accepted: 02/19/2022] [Indexed: 01/10/2023]
Abstract
Objectives The aim of this study was to investigate the effect on standardized uptake value (SUV) measurement variability of the positional relationship between objects of different sizes and the pixel of a positron emission tomography (PET) image. Methods We used a NEMA IEC body phantom comprising six spheres with diameters of 10, 13, 17, 22, 28, and 37 mm. The phantom was filled with 18F solution and contained target-to-background ratios (TBRs) of 2, 4, and 8. The PET data were acquired for 30 min using a SIGNA PET/MR scanner. The PET images were reconstructed with the ordered subsets expectation maximization (OSEM) algorithm with and without point-spread function (PSF) correction (OSEM + PSF + Filter and OSEM + Filter, respectively). A Gaussian filter of 4 mm full width at half maximum was applied in all reconstructions, except for one model (OSEM + PSF + no Filter). The matrix sizes were 128×128, 192×192, 256×256 and 384×384. Reconstruction was performed by shifting the reconstruction center position by 1 mm in the range 0 to 3 mm in the upward or rightward direction for each parameter. For all reconstructed images, the SUVmax of each hot sphere was measured. To investigate the resulting variation in the SUVmax, the coefficient of variation (CV) of each SUVmax was calculated. Results The CV of the SUVmax increased as the matrix size and the diameter of the hot sphere decreased in all reconstruction settings. With PSF correction, the CV of SUVmax increased as the TBR increased except when the TBR was 2. The CV of the SUVmax measured in the OSEM + PSF + no Filter images were larger than those measured in the OSEM + PSF + Filter images. The amount of this increase was higher for smaller spheres and larger matrix sizes and was independent of TBR. Conclusions Shifting the reconstruction center position of the PET image causes variability in SUVmax measurements. To reduce the variability of SUV measurements, it is necessary to use sufficient matrix sizes to satisfy sampling criterion and appropriate filters.
Collapse
Affiliation(s)
- Koji Itagaki
- Corresponding author: Koji Itagaki. Kyoto University Hospital, Division of Clinical Radiology Service, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan. Tel: 075-751-3522;
| | | | | | | | | |
Collapse
|
32
|
Zwezerijnen GJC, Eertink JJ, Ferrández MC, Wiegers SE, Burggraaff CN, Lugtenburg PJ, Heymans MW, de Vet HCW, Zijlstra JM, Boellaard R. Reproducibility of [18F]FDG PET/CT liver SUV as reference or normalisation factor. Eur J Nucl Med Mol Imaging 2023; 50:486-493. [PMID: 36166080 PMCID: PMC9816285 DOI: 10.1007/s00259-022-05977-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/15/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Although visual and quantitative assessments of [18F]FDG PET/CT studies typically rely on liver uptake value as a reference or normalisation factor, consensus or consistency in measuring [18F]FDG uptake is lacking. Therefore, we evaluate the variation of several liver standardised uptake value (SUV) measurements in lymphoma [18F]FDG PET/CT studies using different uptake metrics. METHODS PET/CT scans from 34 lymphoma patients were used to calculate SUVmaxliver, SUVpeakliver and SUVmeanliver as a function of (1) volume-of-interest (VOI) size, (2) location, (3) imaging time point and (4) as a function of total metabolic tumour volume (MTV). The impact of reconstruction protocol on liver uptake is studied on 15 baseline lymphoma patient scans. The effect of noise on liver SUV was assessed using full and 25% count images of 15 lymphoma scans. RESULTS Generally, SUVmaxliver and SUVpeakliver were 38% and 16% higher compared to SUVmeanliver. SUVmaxliver and SUVpeakliver increased up to 31% and 15% with VOI size while SUVmeanliver remained unchanged with the lowest variability for the largest VOI size. Liver uptake metrics were not affected by VOI location. Compared to baseline, liver uptake metrics were 15-18% and 9-18% higher at interim and EoT PET, respectively. SUVliver decreased with larger total MTVs. SUVmaxliver and SUVpeakliver were affected by reconstruction protocol up to 62%. SUVmax and SUVpeak moved 22% and 11% upward between full and 25% count images. CONCLUSION SUVmeanliver was most robust against VOI size, location, reconstruction protocol and image noise level, and is thus the most reproducible metric for liver uptake. The commonly recommended 3 cm diameter spherical VOI-based SUVmeanliver values were only slightly more variable than those seen with larger VOI sizes and are sufficient for SUVmeanliver measurements in future studies. TRIAL REGISTRATION EudraCT: 2006-005,174-42, 01-08-2008.
Collapse
Affiliation(s)
- Gerben J C Zwezerijnen
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | - Maria C Ferrández
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Coreline N Burggraaff
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | | | - Martijn W Heymans
- Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Henrica C W de Vet
- Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Hematology, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Shimizu T, Kim M, Palangka CR, Seki-Soda M, Ogawa M, Takayama Y, Yokoo S. Determination of diagnostic and predictive parameters for vertical mandibular invasion in patients with lower gingival squamous cell carcinoma: A retrospective study. Medicine (Baltimore) 2022; 101:e32206. [PMID: 36626519 PMCID: PMC9750639 DOI: 10.1097/md.0000000000032206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vertical mandibular invasion of lower gingival squamous cell carcinoma (LGSCC) determines the method of resection, which significantly affects the patient's quality of life. Therefore, in mandibular invasion by LGSCC, it is extremely important to monitor progression, specifically whether invasion is limited to the cortical bone or has progressed to the bone marrow. This retrospective study aimed to identify the diagnostic and predictive parameters for mandibular invasion, particularly vertical invasion, to enable appropriate selection of the method of mandibular resection. Of the patients who underwent surgery for LGSCC between 2009 and 2017, 64 were eligible for participation in the study based on tissue microarrays (TMA) from surgical specimens. This study analyzed morphological features using computed tomography (CT), and metabolic characteristics using maximum standardized uptake value (SUVmax), peak value of SUV (SUVpeak), metabolic tumor volume (MTV), and total lesion glycolysis (TLG). Moreover, immunohistochemical analysis of proteins, including parathyroid hormone-related protein (PTHrP), interleukin-6 (IL-6), E-cadherin, and programmed cell death-1 ligand 1 (PD-L1), was performed. Statistical analysis was performed using univariate logistic regression analysis with the forward selection method. The present study showed that MTV (≥2.9 cm3) was an independent diagnostic and predictive factor for positivity of mandibular invasion. Additionally, TLG (≥53.9 bw/cm3) was an independent diagnostic and predictive factor for progression to bone marrow invasion. This study demonstrated that in addition to morphological imaging by CT, the volume-based parameters of MTV and TLG on fluorine-18 fluorodeoxyglucose positron emission tomography were important for predicting pathological mandibular invasion in patients with LGSCC. A more accurate preoperative diagnosis of vertical mandibular invasion would enable the selection of appropriate surgical procedure for mandibular resection.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- * Correspondence: Takahiro Shimizu, Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan (e-mail: )
| | - Mai Kim
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Citra R.A.P. Palangka
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Mai Seki-Soda
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masaru Ogawa
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yu Takayama
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Satoshi Yokoo
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
34
|
Moradi F, Duan H, Song H, Davidzon GA, Chung BI, Thong AEC, Loening AM, Ghanouni P, Sonn G, Iagaru A. 68Ga-PSMA-11 PET/MRI in Patients with Newly Diagnosed Intermediate- or High-Risk Prostate Adenocarcinoma: PET Findings Correlate with Outcomes After Definitive Treatment. J Nucl Med 2022; 63:1822-1828. [PMID: 35512996 DOI: 10.2967/jnumed.122.263897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/22/2022] [Indexed: 01/11/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) PET offers an accuracy superior to other imaging modalities in initial staging of prostate cancer and is more likely to affect management. We examined the prognostic value of 68Ga-PSMA-11 uptake in the primary lesion and presence of metastatic disease on PET in newly diagnosed prostate cancer patients before initial therapy. Methods: In a prospective study from April 2016 to December 2020, 68Ga-PSMA-11 PET/MRI was performed in men with a new diagnosis of intermediate- or high-grade prostate cancer who were candidates for prostatectomy. Patients were followed up after initial therapy for up to 5 y. We examined the Kendall correlation between PET (intense uptake in the primary lesion and presence of metastatic disease) and clinical and pathologic findings (grade group, extraprostatic extension, nodal involvement) relevant for risk stratification, and examined the relationship between PET findings and outcome using Kaplan-Meier analysis. Results: Seventy-three men (age, 64.0 ± 6.3 y) were imaged. Seventy-two had focal uptake in the prostate, and in 20 (27%) PSMA-avid metastatic disease was identified. Uptake correlated with grade group and prostate-specific antigen (PSA). Presence of PSMA metastasis correlated with grade group and pathologic nodal stage. PSMA PET had higher per-patient positivity than nodal dissection in patients with only 5-15 nodes removed (8/41 vs. 3/41) but lower positivity if more than 15 nodes were removed (13/21 vs. 10/21). High uptake in the primary lesion (SUVmax > 12.5, P = 0.008) and presence of PSMA metastasis (P = 0.013) were associated with biochemical failure, and corresponding hazard ratios for recurrence within 2 y (4.93 and 3.95, respectively) were similar to or higher than other clinicopathologic prognostic factors. Conclusion: 68Ga-PSMA-11 PET can risk-stratify patients with intermediate- or high-grade prostate cancer before prostatectomy based on degree of uptake in the prostate and presence of metastatic disease.
Collapse
Affiliation(s)
- Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California;
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Hong Song
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Guido A Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Benjamin I Chung
- Department of Urology, Stanford University, Stanford, California; and
| | - Alan E C Thong
- Department of Urology, Stanford University, Stanford, California; and
| | - Andreas M Loening
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, California
| | - Pejman Ghanouni
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, California
| | - Geoffrey Sonn
- Department of Urology, Stanford University, Stanford, California; and
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
35
|
Duff L, Scarsbrook AF, Mackie SL, Frood R, Bailey M, Morgan AW, Tsoumpas C. A methodological framework for AI-assisted diagnosis of active aortitis using radiomic analysis of FDG PET-CT images: Initial analysis. J Nucl Cardiol 2022; 29:3315-3331. [PMID: 35322380 PMCID: PMC9834376 DOI: 10.1007/s12350-022-02927-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 01/05/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The aim of this study was to explore the feasibility of assisted diagnosis of active (peri-)aortitis using radiomic imaging biomarkers derived from [18F]-Fluorodeoxyglucose Positron Emission Tomography-Computed Tomography (FDG PET-CT) images. METHODS The aorta was manually segmented on FDG PET-CT in 50 patients with aortitis and 25 controls. Radiomic features (RF) (n = 107), including SUV (Standardized Uptake Value) metrics, were extracted from the segmented data and harmonized using the ComBat technique. Individual RFs and groups of RFs (i.e., signatures) were used as input in Machine Learning classifiers. The diagnostic utility of these classifiers was evaluated with area under the receiver operating characteristic curve (AUC) and accuracy using the clinical diagnosis as the ground truth. RESULTS Several RFs had high accuracy, 84% to 86%, and AUC scores 0.83 to 0.97 when used individually. Radiomic signatures performed similarly, AUC 0.80 to 1.00. CONCLUSION A methodological framework for a radiomic-based approach to support diagnosis of aortitis was outlined. Selected RFs, individually or in combination, showed similar performance to the current standard of qualitative assessment in terms of AUC for identifying active aortitis. This framework could support development of a clinical decision-making tool for a more objective and standardized assessment of aortitis.
Collapse
Affiliation(s)
- Lisa Duff
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 8.49b Worsley Building, Clarendon Way, Leeds, LS2 9JT, UK.
- Institute of Medical and Biological Engineering, University of Leeds, Leeds, UK.
| | - Andrew F Scarsbrook
- Leeds Institute of Medical Research - St James's, University of Leeds, Leeds, UK
- Department of Radiology, St. James University Hospital, Leeds, UK
| | - Sarah L Mackie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Biomedical Research Centre, NIHR Leeds, Leeds, UK
| | - Russell Frood
- Leeds Institute of Medical Research - St James's, University of Leeds, Leeds, UK
- Department of Radiology, St. James University Hospital, Leeds, UK
| | - Marc Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 8.49b Worsley Building, Clarendon Way, Leeds, LS2 9JT, UK
- The Leeds Vascular Institute, Leeds General Infirmary, Leeds, UK
| | - Ann W Morgan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 8.49b Worsley Building, Clarendon Way, Leeds, LS2 9JT, UK
- Leeds Teaching Hospitals NHS Trust, Biomedical Research Centre, NIHR Leeds, Leeds, UK
| | - Charalampos Tsoumpas
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 8.49b Worsley Building, Clarendon Way, Leeds, LS2 9JT, UK
- Icahn School of Medicine at Mount Sinai, Biomedical Engineering and Imaging Institute, New York, USA
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center of Groningen, University of Groningen, 9700 RB, Groningen, Netherlands
| |
Collapse
|
36
|
Castillo-Flores S, Gonzalez MR, Bryce-Alberti M, de Souza F, Subhawong TK, Kuker R, Pretell-Mazzini J. PET-CT in the Evaluation of Neoadjuvant/Adjuvant Treatment Response of Soft-tissue Sarcomas: A Comprehensive Review of the Literature. JBJS Rev 2022; 10:01874474-202212000-00003. [PMID: 36639875 DOI: 10.2106/jbjs.rvw.22.00131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
➢ In soft-tissue sarcomas (STSs), the use of positron emission tomography-computed tomography (PET-CT) through a standardized uptake value reduction rate correlates well with histopathological response to neoadjuvant treatment and survival. ➢ PET-CT has shown a better sensitivity to diagnose systemic involvement compared with magnetic resonance imaging and CT; therefore, it has an important role in detecting recurrent systemic disease. However, delaying the use of PET-CT scan, to differentiate tumor recurrence from benign fluorodeoxyglucose uptake changes after surgical treatment and radiotherapy, is essential. ➢ PET-CT limitations such as difficult differentiation between benign inflammatory and malignant processes, inefficient discrimination between benign soft-tissue tumors and STSs, and low sensitivity when evaluating small pulmonary metastases must be of special consideration.
Collapse
Affiliation(s)
- Samy Castillo-Flores
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Marcos R Gonzalez
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mayte Bryce-Alberti
- Medical Student at Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Felipe de Souza
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Ty K Subhawong
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Russ Kuker
- Division of Musculoskeletal Radiology, Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Pretell-Mazzini
- Division of Orthopedic Oncology, Miami Cancer Institute, Baptist Health System South Florida, Plantation, Florida
| |
Collapse
|
37
|
Sun T, Huang S, Jiang Y, Yuan H, Wu J, Liu C, Zhang X, Tang Y, Ben X, Tang J, Zhou H, Zhang D, Xie L, Chen G, Zhao Y, Wang S, Xu H, Qiao G. Dynamic alteration in SULmax predicts early pathological tumor response and short-term prognosis in non-small cell lung cancer treated with neoadjuvant immunochemotherapy. Front Bioeng Biotechnol 2022; 10:1010672. [PMID: 36277407 PMCID: PMC9582780 DOI: 10.3389/fbioe.2022.1010672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction: Biomarkers predicting tumor response to neoadjuvant immunochemotherapy in non-small cell lung cancer (NSCLC) are still lacking despite great efforts. We aimed to assess the effectiveness of the immune PET Response Criteria in Solid Tumors via SULmax (iPERCIST-max) in predicting tumor response to neoadjuvant immunochemotherapy and short-term survival in locally advanced NSCLC. Methods: In this prospective cohort study, we calculated SULmax, SULpeak, metabolic tumor volume (MTV), total lesion glycolysis (TLG) and their dynamic percentage changes in a training cohort. We then investigated the correlation between alterations in these parameters and pathological tumor responses. Subsequently, iPERCIST-max defined by the proportional changes in the SULmax response (△SULmax%) was constructed and internally validated using a time-dependent receiver operating characteristic (ROC) curve and the area under the curve (AUC) value. A prospective cohort from the Sun Yat-Sen University Cancer Center (SYSUCC) was also included for external validation. The relationship between the iPERCIST-max responsiveness and event-free survival in the training cohort was also investigated. Results: Fifty-five patients with NSCLC were included in this study from May 2019 to December 2021. Significant alterations in post-treatment SULmax (p < 0.001), SULpeak (p < 0.001), SULmean (p < 0.001), MTV (p < 0.001), TLG (p < 0.001), and tumor size (p < 0.001) were observed compared to baseline values. Significant differences in SULpeak, SULmax, and SULmean between major pathological response (mPR) and non-mPR statuses were observed. The optimal cutoff values of the SULmax response rate were -70.0% and -88.0% using the X-tile software. The univariate and multivariate binary logistic regression showed that iPERCIST-max is the only significant key predictor for mPR status [OR = 84.0, 95% confidence interval (CI): 7.84-900.12, p < 0.001]. The AUC value for iPERCIST-max was 0.896 (95% CI: 0.776-1.000, p < 0.001). Further, external validation showed that the AUC value for iPERCIST-max in the SYSUCC cohort was 0.889 (95% CI: 0.698-1.000, p = 0.05). Significantly better event-free survival (EFS) in iPERCIST-max responsive disease (31.5 months, 95% CI 27.9-35.1) than that in iPERCIST-max unresponsive disease (22.2 months, 95% CI: 17.3-27.1 months, p = 0.024) was observed. Conclusion: iPERCIST-max could better predict both early pathological tumor response and short-term prognosis of NSCLC treated with neoadjuvant immunochemotherapy than commonly used criteria. Furthermore, large-scale prospective studies are required to confirm the generalizability of our findings.
Collapse
Affiliation(s)
- Taotao Sun
- Department of Nuclear Medicine and PET/CT-MRI Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Nuclear Medicine, WeiLun PET Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shujie Huang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yongluo Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Yuan
- Department of Nuclear Medicine, WeiLun PET Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junhan Wu
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Chao Liu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaochun Zhang
- Department of Nuclear Medicine, WeiLun PET Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yong Tang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaosong Ben
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiming Tang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haiyu Zhou
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dongkun Zhang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liang Xie
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Gang Chen
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yumo Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuxia Wang
- Department of Nuclear Medicine, WeiLun PET Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Braune A, Oehme L, Freudenberg R, Hofheinz F, van den Hoff J, Kotzerke J, Hoberück S. Comparison of image quality and spatial resolution between 18F, 68Ga, and 64Cu phantom measurements using a digital Biograph Vision PET/CT. EJNMMI Phys 2022; 9:58. [PMID: 36064989 PMCID: PMC9445107 DOI: 10.1186/s40658-022-00487-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND PET nuclides can have a considerable influence on the spatial resolution and image quality of PET/CT scans, which can influence diagnostics in oncology, for example. The individual impact of the positron energy of 18F, 68Ga, and 64Cu on spatial resolution and image quality was compared for PET/CT scans acquired using a clinical, digital scanner. METHODS A Jaszczak phantom and a NEMA PET body phantom were filled with 18F-FDG, 68Ga-HCl, or 64Cu-HCl, and PET/CT scans were performed on a Siemens Biograph Vision. Acquired images were analyzed regarding spatial resolution and image quality (recovery coefficients (RC), coefficient of variation within the background, contrast recovery coefficient (CRC), contrast-noise ratio (CNR), and relative count error in the lung insert). Data were compared between scans with different nuclides. RESULTS We found that image quality was comparable between 18F-FDG and 64Cu-HCl PET/CT measurements featuring similar maximal endpoint energies of the positrons. In comparison, RC, CRC, and CNR were degraded in 68Ga-HCl data despite similar count rates. In particular, the two smallest spheres of 10 mm and 13 mm diameter revealed lower RC, CRC, and CNR values. The spatial resolution was similar between 18F-FDG and 64Cu-HCl but up to 18% and 23% worse compared with PET/CT images of the NEMA PET body phantom filled with 68Ga-HCl. CONCLUSIONS The positron energy of the PET nuclide influences the spatial resolution and image quality of a digital PET/CT scan. The image quality and spatial resolution of 68Ga-HCl PET/CT images were worse than those of 18F-FDG or 64Cu-HCl despite similar count rates.
Collapse
Affiliation(s)
- Anja Braune
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| | - Liane Oehme
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Robert Freudenberg
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Frank Hofheinz
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jörg van den Hoff
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.,PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Nuclear Medicine, Medizinische Fakultat Carl Gustav Carus, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Sebastian Hoberück
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.,Department of Nuclear Medicine, Helios Klinikum Erfurt, Erfurt, Germany
| |
Collapse
|
39
|
Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment. Int J Mol Sci 2022; 23:ijms23169394. [PMID: 36012657 PMCID: PMC9409366 DOI: 10.3390/ijms23169394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Evaluation of cancer therapy with imaging is crucial as a surrogate marker of effectiveness and survival. The unique response patterns to therapy with immune-checkpoint inhibitors have facilitated the revision of response evaluation criteria using FDG-PET, because the immune response recalls reactive cells such as activated T-cells and macrophages, which show increased glucose metabolism and apparent progression on morphological imaging. Cellular metabolism and function are critical determinants of the viability of active cells in the tumor microenvironment, which would be novel targets of therapies, such as tumor immunity, metabolism, and genetic mutation. Considering tumor heterogeneity and variation in therapy response specific to the mechanisms of therapy, appropriate response evaluation is required. Radiomics approaches, which combine objective image features with a machine learning algorithm as well as pathologic and genetic data, have remarkably progressed over the past decade, and PET radiomics has increased quality and reliability based on the prosperous publications and standardization initiatives. PET and multimodal imaging will play a definitive role in personalized therapeutic strategies by the precise monitoring in future cancer therapy.
Collapse
|
40
|
Harmonization based on quantitative analysis of standardized uptake value variations across PET/CT scanners: a multicenter phantom study. Nucl Med Commun 2022; 43:1004-1014. [PMID: 35836388 DOI: 10.1097/mnm.0000000000001598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES This study aimed to measure standardized uptake value (SUV) variations across different PET/computed tomography (CT) scanners to harmonize quantification across systems. METHODS We acquired images using the National Electrical Manufacturers Association International Electrotechnical Commission phantom from three PET/CT scanners operated using routine imaging protocols at each site. The SUVs of lesions were assessed in the presence of reference values by a digital reference object (DRO) and recommendations by the European Association of Nuclear Medicine (EANM/EARL) to measure inter-site variations. For harmonization, Gaussian filters with tuned full width at half maximum (FWHM) values were applied to images to minimize differences in SUVs between reference and images. Inter-site variation of SUVs was evaluated in both pre- and postharmonization situations. Test-retest analysis was also carried out to evaluate repeatability. RESULTS SUVs from different scanners became significantly more consistent, and inter-site differences decreased for SUVmean, SUVmax and SUVpeak from 17.3, 20.7, and 15.5% to 4.8, 4.7, and 2.7%, respectively, by harmonization (P values <0.05 for all). The values for contrast-to-noise ratio in the smallest lesion of the phantom verified preservation of image quality following harmonization (>2.8%). CONCLUSIONS Harmonization significantly lowered variations in SUV measurements across different PET/CT scanners, improving reproducibility while preserving image quality.
Collapse
|
41
|
Parghane RV, Basu S. PET-CTBased Quantitative Parameters for Assessment of Treatment Response and Disease Activity in Cancer and Noncancerous Disorders. PET Clin 2022; 17:465-478. [PMID: 35717102 DOI: 10.1016/j.cpet.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The various semiquantitative and quantitative PET-CT parameters provide measurement of disease activity and assessment of treatment response in the PET-CT studies. These include standardized uptake value (SUV), metabolic tumor volume (MTV) and total lesion glycolysis (TLG), and total metabolic tumor volume (TMTV). Thresholding and adaptive thresholding methods are commonly used algorithms for the evaluation of global disease activity. Readily available commercial software frequently in-built with the current generation PET-CT scanners for providing easy, less time consuming, highly reproducible, and more accurate measurement of global disease activity on PET-CT imaging in evaluation of malignant as well as benign disorders.
Collapse
Affiliation(s)
- Rahul V Parghane
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
42
|
De Luca GMR, Habraken JBA. Method to determine the statistical technical variability of SUV metrics. EJNMMI Phys 2022; 9:40. [PMID: 35666316 PMCID: PMC9170854 DOI: 10.1186/s40658-022-00470-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Standardized Uptake Value (SUV) Max, SUVMean, and SUVPeak are metrics used to quantify positron emission tomography (PET) images. In order to assess the significance of a change in these metrics for diagnostic purposes, it is relevant to know their variation. The sources of variation can be biological or technical. In this study, we present a method to determine the statistical technical variation of SUV in PET images. RESULTS This method was tested on a NEMA quality phantom with spheres of various diameters with a full-length acquisition time of 150 s per bed position and foreground-to-background activity ratio of F18-2-fluoro-2-deoxy-D-glucose (FDG) of 10:1. Our method divides the 150 s acquisition into subsets with statistically independent frames of shorter reconstruction length. SUVMax, Mean and Peak were calculated for each reconstructed image in a subset. The coefficient of variation of SUV within each subset has been used to estimate the expected coefficient of variation at 150 s reconstruction length. We report the largest coefficient of variation of the SUV metrics for the smallest sphere and the smallest variation for the largest sphere. The expected variation at 150 s reconstruction length does not exceed 6% for the smallest sphere and 2% for the largest sphere. CONCLUSIONS With the presented method, we aim to determine the statistical technical variation of SUV. The method enables the evaluation of the effect of SUV metric choice (Max, Mean, Peak) and lesion size on the technical variation and, therefore, to evaluate its relevance on the total variation of the SUV value between clinical studies.
Collapse
Affiliation(s)
- Giulia M R De Luca
- Department of Medical Physics, St. Antonius Hospital, Nieuwegein, The Netherlands.
| | - Jan B A Habraken
- Department of Medical Physics, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
43
|
Zhang Y, Hu P, He Y, Yu H, Tan H, Liu G, Gu J, Shi H. Ultrafast 30-s total-body PET/CT scan: a preliminary study. Eur J Nucl Med Mol Imaging 2022; 49:2504-2513. [PMID: 35578037 DOI: 10.1007/s00259-022-05838-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/07/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE The aim of this study is to explore the diagnostic value of the images obtained in ultrafast 30-s acquisition time by the total-body PET/CT (18F-FDG injection dose of about 3.7 MBq/kg), and to evaluate whether they can meet the requirements of clinical diagnosis or not. METHODS This retrospective study explored the clinical value of ultrafast 30-s 18F-FDG total-body PET/CT in 88 oncology patients, using the post-surgical pathological diagnosis as the reference standard. The data were acquired over 300 s and reconstructed using all 300-s data (G300) and only the initial 30 s (G30). Two readers independently assessed all images qualitatively and quantitatively. The diagnostic performance was compared between G300 and G30. RESULTS The G300 average qualitative score was higher than G30 (P < 0.001). G300 and G30 also differed quantitatively in the liver and mediastinum SUVmax, SD, and SNR (all P < 0.001), but had similar sensitivities (89.09% vs. 86.36%, P = 0.250). The G300 group had higher accuracy (79.73%) and a larger area under the curve (0.709) than G30 (77.70% and 0.695, respectively; all P > 0.05). CONCLUSION The 30-s total-body PET/CT could meet clinical diagnostic requirements for malignant tumors in patients intolerant to prolonged horizontal positioning.
Collapse
Affiliation(s)
- Yiqiu Zhang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Pengcheng Hu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Yibo He
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Haojun Yu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China.,Institute of Nuclear Medicine, Fudan University, Shanghai, China.,Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jianying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai, China. .,Institute of Nuclear Medicine, Fudan University, Shanghai, China. .,Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
44
|
Thomas MA, Meier JG, Mawlawi OR, Sun P, Pan T. Impact of acquisition time and misregistration with CT on data-driven gated PET. Phys Med Biol 2022; 67:10.1088/1361-6560/ac5f73. [PMID: 35313286 PMCID: PMC9128538 DOI: 10.1088/1361-6560/ac5f73] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/21/2022] [Indexed: 11/11/2022]
Abstract
Objective. Data-driven gating (DDG) can address patient motion issues and enhance PET quantification but suffers from increased image noise from utilization of <100% of PET data. Misregistration between DDG-PET and CT may also occur, altering the potential benefits of gating. Here, the effects of PET acquisition time and CT misregistration were assessed with a combined DDG-PET/DDG-CT technique.Approach. In the primary PET bed with lesions of interest and likely respiratory motion effects, PET acquisition time was extended to 12 min and a low-dose cine CT was acquired to enable DDG-CT. Retrospective reconstructions were created for both non-gated (NG) and DDG-PET using 30 s to 12 min of PET data. Both the standard helical CT and DDG-CT were used for attenuation correction of DDG-PET data. SUVmax, SUVpeak, and CNR were compared for 45 lesions in the liver and lung from 27 cases.Main results. For both NG-PET (p= 0.0041) and DDG-PET (p= 0.0028), only the 30 s acquisition time showed clear SUVmaxbias relative to the 3 min clinical standard. SUVpeakshowed no bias at any change in acquisition time. DDG-PET alone increased SUVmaxby 15 ± 20% (p< 0.0001), then was increased further by an additional 15 ± 29% (p= 0.0007) with DDG-PET/CT. Both 3 min and 6 min DDG-PET had lesion CNR statistically equivalent to 3 min NG-PET, but then increased at 12 min by 28 ± 48% (p= 0.0022). DDG-PET/CT at 6 min had comparable counts to 3 min NG-PET, but significantly increased CNR by 39 ± 46% (p< 0.0001).Significance. 50% counts DDG-PET did not lead to inaccurate or biased SUV-increased SUV resulted from gating. Improved registration from DDG-CT was equally as important as motion correction with DDG-PET for increasing SUV in DDG-PET/CT. Lesion detectability could be significantly improved when DDG-PET used equivalent counts to NG-PET, but only when combined with DDG-CT in DDG-PET/CT.
Collapse
Affiliation(s)
- M. Allan Thomas
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX 77030
| | - Joseph G. Meier
- Department of Medical Physics, University of Wisconsin, Madison, WI 53726
| | - Osama R. Mawlawi
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX 77030
| | - Peng Sun
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX 77030
| | - Tinsu Pan
- Department of Imaging Physics, UT MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
45
|
Zirakchian Zadeh M, Ayubcha C, Raynor WY, Werner TJ, Alavi A. A review of different methods used for quantification and assessment of FDG-PET/CT in multiple myeloma. Nucl Med Commun 2022; 43:378-391. [PMID: 35102074 DOI: 10.1097/mnm.0000000000001528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The quantification of positron emission tomography/computed tomography (PET/CT) in multiple myeloma (MM) is challenging. Different methods of PET/CT quantification for assessment of fluorodeoxyglucose (FDG) uptake in myeloma patients have been suggested. This is the first review article that focuses on the advantages and disadvantages of each approach. Use of the maximum standardized uptake value (SUVmax) showed some promise in prognostic stratification of MM patients. However, it is affected by noise and time of flight and is subject to high variability. Volumetric PET metrics such as total lesion glycolysis and metabolic tumor volume are other proposed approaches. The high number of osteolytic lesions in MM patients makes this approach difficult in clinical practice. In addition, evaluation of small focal lesions is subject to partial volume correction. CT-based segmentation for assessment of FDG radiotracer is recently introduced. The methodologies are highly reproducible, but the clinical values of the approaches are unclear and still under investigation. We also discuss the Italian Myeloma criteria for PET Use (IMPeTUs), which is a qualitative approach, as a point of comparison. The reproducibility of IMPeTUs depends heavily on the level of user experience. We recommend further studies for assessing the prognostic significance of CT-threshold approaches in the assessment of MM patients.
Collapse
Affiliation(s)
| | | | - William Y Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, Pennsylvania, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Pennsylvania, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
46
|
Driscoll B, Shek T, Vines D, Sun A, Jaffray D, Yeung I. Phantom Validation of a Conservation of Activity-Based Partial Volume Correction Method for Arterial Input Function in Dynamic PET Imaging. Tomography 2022; 8:842-857. [PMID: 35314646 PMCID: PMC8938778 DOI: 10.3390/tomography8020069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
Dynamic PET (dPET) imaging can be utilized to perform kinetic modelling of various physiologic processes, which are exploited by the constantly expanding range of targeted radiopharmaceuticals. To date, dPET remains primarily in the research realm due to a number of technical challenges, not least of which is addressing partial volume effects (PVE) in the input function. We propose a series of equations for the correction of PVE in the input function and present the results of a validation study, based on a purpose built phantom. 18F-dPET experiments were performed using the phantom on a set of flow tubes representing large arteries, such as the aorta (1" 2.54 cm ID), down to smaller vessels, such as the iliac arteries and veins (1/4" 0.635 cm ID). When applied to the dPET experimental images, the PVE correction equations were able to successfully correct the image-derived input functions by as much as 59 ± 35% in the presence of background, which resulted in image-derived area under the curve (AUC) values within 8 ± 9% of ground truth AUC. The peak heights were similarly well corrected to within 9 ± 10% of the scaled DCE-CT curves. The same equations were then successfully applied to correct patient input functions in the aorta and internal iliac artery/vein. These straightforward algorithms can be applied to dPET images from any PET-CT scanner to restore the input function back to a more clinically representative value, without the need for high-end Time of Flight systems or Point Spread Function correction algorithms.
Collapse
Affiliation(s)
- Brandon Driscoll
- Quantitative Imaging for Personalized Cancer Medicine (QIPCM)—Techna Institute, University Health Network, Toronto, ON M5G 2C4, Canada; (T.S.); (D.J.); (I.Y.)
- Correspondence:
| | - Tina Shek
- Quantitative Imaging for Personalized Cancer Medicine (QIPCM)—Techna Institute, University Health Network, Toronto, ON M5G 2C4, Canada; (T.S.); (D.J.); (I.Y.)
| | - Douglass Vines
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; (D.V.); (A.S.)
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Alex Sun
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; (D.V.); (A.S.)
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - David Jaffray
- Quantitative Imaging for Personalized Cancer Medicine (QIPCM)—Techna Institute, University Health Network, Toronto, ON M5G 2C4, Canada; (T.S.); (D.J.); (I.Y.)
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; (D.V.); (A.S.)
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ivan Yeung
- Quantitative Imaging for Personalized Cancer Medicine (QIPCM)—Techna Institute, University Health Network, Toronto, ON M5G 2C4, Canada; (T.S.); (D.J.); (I.Y.)
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; (D.V.); (A.S.)
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
47
|
Cheung AHY, Wu VWC, Cheung ALY, Cai J. Respiratory 4D-Gating F-18 FDG PET/CT Scan for Liver Malignancies: Feasibility in Liver Cancer Patient and Tumor Quantitative Analysis. Front Oncol 2022; 12:789506. [PMID: 35223472 PMCID: PMC8864173 DOI: 10.3389/fonc.2022.789506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/12/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose To evaluate the potential clinical role and effectiveness of respiratory 4D-gating F-18 FDG PET/CT scan for liver malignancies, relative to routine (3D) F-18 FDG PET/CT scan. Materials and Methods This study presented a prospective clinical study of 16 patients who received F-18 FDG PET/CT scan for known or suspected malignant liver lesions. Ethics approvals were obtained from the ethics committees of the Hong Kong Baptist Hospital and The Hong Kong Polytechnic University. Liver lesions were compared between the gated and ungated image sets, in terms of 1) volume measurement of PET image, 2) accuracy of maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), and 3) accuracy of total lesion glycoses (TLG). Statistical analysis was performed by using a two-tailed paired Student t-test and Pearson correlation test. Results The study population consisted of 16 patients (9 males and 7 females; mean age of 65) with a total number of 89 lesions. The SUVmax and SUVmean measurement of the gated PET images was more accurate than that of the ungated PET images, compared to the static reference images. An average of 21.48% (p < 0.001) reduction of the tumor volume was also observed. The SUVmax and SUVmean of the gated PET images were improved by 19.81% (p < 0.001) and 25.53% (p < 0.001), compared to the ungated PET images. Conclusions We have demonstrated the feasibility of implementing 4D PET/CT scan for liver malignancies in a prospective clinical study. The 4D PET/CT scan for liver malignancies could improve the quality of PET image by improving the SUV accuracy of the lesions and reducing image blurring. The improved accuracy in the classification and identification of liver tumors with 4D PET image would potentially lead to its increased utilization in target delineation of GTV, ITV, and PTV for liver radiotherapy treatment planning in the future.
Collapse
Affiliation(s)
- Anson H Y Cheung
- Department of Health Technology & Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China.,Radiotherapy and Oncology Department, Hong Kong Baptist Hospital, Hong Kong, Hong Kong SAR, China
| | - Vincent W C Wu
- Department of Health Technology & Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Andy L Y Cheung
- Department of Health Technology & Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China.,Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Jing Cai
- Department of Health Technology & Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
48
|
Ruan W, Liu F, Sun X, Hu F, Wu T, Zhang Y, Lan X. Evaluating two respiratory correction methods for abdominal PET/MRI imaging. EJNMMI Phys 2022; 9:5. [PMID: 35099646 PMCID: PMC8804027 DOI: 10.1186/s40658-022-00430-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background To evaluate two respiratory correction methods for abdominal PET/MRI images and further to analyse the effects on standard uptake values (SUVs) of respiratory motion correction, 17 patients with 25 abdominal lesions on 18F-FDG PET/CT were scanned with PET/MRI. PET images were reconstructed using end-expiratory respiratory gating and multi-bin respiratory gating. Meanwhile, full data and the first 3 min and 20 s of data acquired both without respiratory gating were reconstructed for evaluation. Five parameters, including the SUVmax and SUVmean in the lesions, the SUVmean and standard deviation (SD) in the background, and the signal-to-noise ratio (SNR), were calculated and used for statistical comparisons. The differences in multi-bin respiratory gating and reconstruction of full data, relative to the reconstruction of the first 3 min and 20 s of data acquired, were calculated. Results Compared with PET/CT, the longer scanning time of abdominal PET/MRI makes respiratory motion correction necessary. The multi-bin respiratory gating correction could reduce the PET image blur and increase the SUVmax (11.98%) and SUVmean (13.12%) of the lesions significantly (p = 0.00), which was much more effective than end-expiratory respiratory gating for abdominal PET/MRI. The added value of SUVmax caused by respiratory motion correction has no significant difference compared with that caused by count loss with the correction (p = 0.39), which was rarely reported by previous studies. Conclusion Based on the current parameters, the method of multi-bin respiratory gating was more effective for respiratory motion correction in abdominal PET/MRI in comparisons with the method of end-respiratory gating. However, the increased noise in gated images, due to the fact that PET data get discarded, is partly responsible for the increase in SUVmax.
Collapse
Affiliation(s)
- Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Fang Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xun Sun
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | | | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
49
|
Akamatsu G, Shimada N, Matsumoto K, Daisaki H, Suzuki K, Watabe H, Oda K, Senda M, Terauchi T, Tateishi U. New standards for phantom image quality and SUV harmonization range for multicenter oncology PET studies. Ann Nucl Med 2022; 36:144-161. [PMID: 35029817 DOI: 10.1007/s12149-021-01709-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/05/2021] [Indexed: 11/01/2022]
Abstract
Not only visual interpretation for lesion detection, staging, and characterization, but also quantitative treatment response assessment are key roles for 18F-FDG PET in oncology. In multicenter oncology PET studies, image quality standardization and SUV harmonization are essential to obtain reliable study outcomes. Standards for image quality and SUV harmonization range should be regularly updated according to progress in scanner performance. Accordingly, the first aim of this study was to propose new image quality reference levels to ensure small lesion detectability. The second aim was to propose a new SUV harmonization range and an image noise criterion to minimize the inter-scanner and intra-scanner SUV variabilities. We collected a total of 37 patterns of images from 23 recent PET/CT scanner models using the NEMA NU2 image quality phantom. PET images with various acquisition durations of 30-300 s and 1800 s were analyzed visually and quantitatively to derive visual detectability scores of the 10-mm-diameter hot sphere, noise-equivalent count (NECphantom), 10-mm sphere contrast (QH,10 mm), background variability (N10 mm), contrast-to-noise ratio (QH,10 mm/N10 mm), image noise level (CVBG), and SUVmax and SUVpeak for hot spheres (10-37 mm diameters). We calculated a reference level for each image quality metric, so that the 10-mm sphere can be visually detected. The SUV harmonization range and the image noise criterion were proposed with consideration of overshoot due to point-spread function (PSF) reconstruction. We proposed image quality reference levels as follows: QH,10 mm/N10 mm ≥ 2.5 and CVBG ≤ 14.1%. The 10th-90th percentiles in the SUV distributions were defined as the new SUV harmonization range. CVBG ≤ 10% was proposed as the image noise criterion, because the intra-scanner SUV variability significantly depended on CVBG. We proposed new image quality reference levels to ensure small lesion detectability. A new SUV harmonization range (in which PSF reconstruction is applicable) and the image noise criterion were also proposed for minimizing the SUV variabilities. Our proposed new standards will facilitate image quality standardization and SUV harmonization of multicenter oncology PET studies. The reliability of multicenter oncology PET studies will be improved by satisfying the new standards.
Collapse
Affiliation(s)
- Go Akamatsu
- National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Naoki Shimada
- Cancer Institute Hospital, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan.
| | - Keiichi Matsumoto
- Kyoto College of Medical Science, 1-3 Imakita, Oyamahigashi-cho, Sonobe-cho, Nantan, Kyoto, 622-0041, Japan
| | - Hiromitsu Daisaki
- Gunma Prefectural College of Health Sciences, 323-1 Kamioki-machi, Maebashi, Gunma, 371-0052, Japan
| | - Kazufumi Suzuki
- Dokkyo Medical University Hospital, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi, 321-0293, Japan
| | - Hiroshi Watabe
- Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Keiichi Oda
- Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido, 006-8585, Japan
| | - Michio Senda
- Kobe City Medical Center General Hospital, 2-1-1 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Takashi Terauchi
- Cancer Institute Hospital, 3-8-31 Ariake, Koto, Tokyo, 135-8550, Japan
| | - Ukihide Tateishi
- Tokyo Medical and Dental University School of Medicine, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
50
|
Fournier L, de Geus-Oei LF, Regge D, Oprea-Lager DE, D’Anastasi M, Bidaut L, Bäuerle T, Lopci E, Cappello G, Lecouvet F, Mayerhoefer M, Kunz WG, Verhoeff JJC, Caruso D, Smits M, Hoffmann RT, Gourtsoyianni S, Beets-Tan R, Neri E, deSouza NM, Deroose CM, Caramella C. Twenty Years On: RECIST as a Biomarker of Response in Solid Tumours an EORTC Imaging Group - ESOI Joint Paper. Front Oncol 2022; 11:800547. [PMID: 35083155 PMCID: PMC8784734 DOI: 10.3389/fonc.2021.800547] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Response evaluation criteria in solid tumours (RECIST) v1.1 are currently the reference standard for evaluating efficacy of therapies in patients with solid tumours who are included in clinical trials, and they are widely used and accepted by regulatory agencies. This expert statement discusses the principles underlying RECIST, as well as their reproducibility and limitations. While the RECIST framework may not be perfect, the scientific bases for the anticancer drugs that have been approved using a RECIST-based surrogate endpoint remain valid. Importantly, changes in measurement have to meet thresholds defined by RECIST for response classification within thus partly circumventing the problems of measurement variability. The RECIST framework also applies to clinical patients in individual settings even though the relationship between tumour size changes and outcome from cohort studies is not necessarily translatable to individual cases. As reproducibility of RECIST measurements is impacted by reader experience, choice of target lesions and detection/interpretation of new lesions, it can result in patients changing response categories when measurements are near threshold values or if new lesions are missed or incorrectly interpreted. There are several situations where RECIST will fail to evaluate treatment-induced changes correctly; knowledge and understanding of these is crucial for correct interpretation. Also, some patterns of response/progression cannot be correctly documented by RECIST, particularly in relation to organ-site (e.g. bone without associated soft-tissue lesion) and treatment type (e.g. focal therapies). These require specialist reader experience and communication with oncologists to determine the actual impact of the therapy and best evaluation strategy. In such situations, alternative imaging markers for tumour response may be used but the sources of variability of individual imaging techniques need to be known and accounted for. Communication between imaging experts and oncologists regarding the level of confidence in a biomarker is essential for the correct interpretation of a biomarker and its application to clinical decision-making. Though measurement automation is desirable and potentially reduces the variability of results, associated technical difficulties must be overcome, and human adjudications may be required.
Collapse
Affiliation(s)
- Laure Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Université de Paris, Assistance Publique–Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Department of Radiology, Paris Cardiovascular Research Center (PARCC) Unité Mixte de Recherche (UMRS) 970, Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| | - Lioe-Fee de Geus-Oei
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Daniele Regge
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Daniela-Elena Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers [Vrije Universiteit (VU) University], Amsterdam, Netherlands
| | - Melvin D’Anastasi
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Medical Imaging Department, Mater Dei Hospital, University of Malta, Msida, Malta
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Tobias Bäuerle
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Egesta Lopci
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine Unit, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) – Humanitas Research Hospital, Milan, Italy
| | - Giovanni Cappello
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Frederic Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marius Mayerhoefer
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang G. Kunz
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Joost J. C. Verhoeff
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Damiano Caruso
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Marion Smits
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
- Brain Tumour Centre, Erasmus Medical Centre (MC) Cancer Institute, Rotterdam, Netherlands
| | - Ralf-Thorsten Hoffmann
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl-Gustav-Carus Technical University Dresden, Dresden, Germany
| | - Sofia Gourtsoyianni
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Areteion Hospital, Athens, Greece
| | - Regina Beets-Tan
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- School For Oncology and Developmental Biology (GROW) School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Emanuele Neri
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Diagnostic and Interventional Radiology, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, United States
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Caroline Caramella
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Radiology Department, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph Centre International des Cancers Thoraciques, Université Paris-Saclay, Le Plessis-Robinson, France
| |
Collapse
|